AU2005276634B2 - Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain - Google Patents

Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain Download PDF

Info

Publication number
AU2005276634B2
AU2005276634B2 AU2005276634A AU2005276634A AU2005276634B2 AU 2005276634 B2 AU2005276634 B2 AU 2005276634B2 AU 2005276634 A AU2005276634 A AU 2005276634A AU 2005276634 A AU2005276634 A AU 2005276634A AU 2005276634 B2 AU2005276634 B2 AU 2005276634B2
Authority
AU
Australia
Prior art keywords
pain
induced
use according
treatment
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2005276634A
Other versions
AU2005276634A1 (en
Inventor
Bettina Beyreuther
Thomas Stohr
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UCB Pharma GmbH
Original Assignee
UCB Pharma GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from EP04020402A external-priority patent/EP1629839A1/en
Application filed by UCB Pharma GmbH filed Critical UCB Pharma GmbH
Publication of AU2005276634A1 publication Critical patent/AU2005276634A1/en
Assigned to UCB PHARMA GMBH reassignment UCB PHARMA GMBH Amend patent request/document other than specification (104) Assignors: SCHWARZ PHARMA AG
Application granted granted Critical
Publication of AU2005276634B2 publication Critical patent/AU2005276634B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids

Description

WO 2006/021412 PCT/EP2005/009067 Novel Use of Peptide Compounds for Treating Bone Cancer Pain, Chemotherapy- and Nucleoside-Induced Pain Description The present invention is directed to the use of a class of peptide compounds for treating tumor pain, in particular bone cancer pain, for treating 5 chemotherapy-induced pain and for treating nucleoside-induced pain. Certain peptides are known to exhibit central nervous system (CNS) activity and are useful in the treatment of epilepsy and other CNS disorders. These peptides which are described in the U.S. Patent No. 5,378,729 have the 10 Formula (1a): R2 R- NH-[-C-CNH-]n-C-R1 15 11 1 11 o o
R
3 20 Formula (la) wherein R is hydrogen, lower alkyl, lower alkenyl, lower alkynyl, aryl, aryl lower alkyl, 25 heterocyclic, heterocyclic lower alkyl, lower alkyl heterocyclic, lower cycloalkyl, lower cycloalkyl lower alkyl, and R is unsubstituted or is substituted with at least one electron withdrawing group or electron donating group; 30 R 1 is hydrogen or lower alkyl, lower alkenyl, lower alkynyl, aryl lower alkyl, aryl, heterocyclic lower alkyl, heterocyclic, lower cycloalkyl, lower cycloalkyl lower alkyl, each unsubstituted or substituted with an electron donating WO 2006/021412 PCT/EP2005/009067 -2 group or an electron withdrawing group; and
R
2 and R 3 are independently hydrogen, lower alkyl, lower alkenyl, lower alkynyl, aryl lower alkyl, aryl, heterocyclic, heterocyclic lower alkyl, lower alkyl heterocyclic, lower cycloalkyl, lower cycloalkyl lower alkyl, or Z-Y 5 wherein R 2 and R 3 may be unsubstituted or substituted with at least one electron withdrawing group or electron donating group; Z is 0, S, S(O)., NR 4 , PR 4 or a chemical bond; 10 Y is hydrogen, lower alkyl, aryl, aryl lower alkyl, lower alkenyl, lower alkynyl, halo, heterocyclic, heterocyclic lower alkyl, and Y may be unsubstituted or substituted with an electron donating group or an electron withdrawing group, provided that when Y is halo, Z is a chemical bond, or 15 ZY taken together is NR 4
NR
5
R
7 , NR 4
OR
5 , ONR 4
R
7 , OPR 4
R
5 , PR 4
OR
5 ,
SNR
4
R
7 , NR 4
SR
7 , SPR 4
R
5 or PR 4
SR
7 , NR 4 PRsR 6 or PR 4 NRsR 7 ,
NR
4
C-R
5 , SCR 5 , NR 4 C-ORs, SC-OR5; 20 || || || || 0 0 0 0
R
4 , R5 and R 6 are independently hydrogen, lower alkyl, aryl, aryl lower alkyl, lower alkenyl, or lower alkynyl, wherein R 4 , R 5 and R 6 may be unsubstituted 25 or substituted with an electron withdrawing group or an electron donating group; and
R
7 is R 6 or COORa or COR 8 ; 30 R 8 is hydrogen or lower alkyl, or aryl lower alkyl, and the aryl or alkyl group may be unsubstituted or substituted with an electron withdrawing group or an electron donating group; and n is 1-4; and 35 a is 1-3.
WO 2006/021412 PCT/EP2005/009067 -3 U.S. Patent No. 5,773,475 also discloses additional compounds useful for treating CNS disorders. These compounds are N-benzyl-2-amino-3 methoxy-propionamide having the Formula (Ila): 5 H H H I I I Ar-CHz-N-C-C-N-C-R 1 10 11 1 11 0 CH 2 0
R
3 15 Formula (Ila) wherein Ar is aryl which is unsubstituted or substituted with halo; R 3 is lower alkoxy; and R 1 is methyl. 20 The patents US 5.378.729 and US 5.773.475 are hereby incorporated by reference. However, neither of these patents describes the use of these compounds for treating tumor pain, in particular bone cancer pain, for treating chemotherapy-induced pain and for treating nucleoside-induced 25 pain. WO 02/074297 relates to the use of a compound according to Formula (Ila) wherein Ar is phenyl which may be substituted by at least one halo, R 3 is lower alkoxy containing 1-3 carbon atoms and R 1 is methyl for the 30 preparation of pharmaceutical compositions useful for the treatment of allodynia related to peripheral neuropathic pain. WO 02/074784 relates to the use of a compound having Formula (la) or/and Formula (Ila) showing antinociceptive properties for treating different types and symptoms of acute and chronic pain, especially non neuropathic WO 2006/021412 PCT/EP2005/009067 -4 inflammatory pain, e.g. rheumatoid arthritic pain or/and secondary inflammatory osteo-arthritic pain. At present no analgesic exists which is highly potent in various pain syndromes. Different mechanisms leading to inflammatory or neuropathic pain make it difficult to identify compounds which have general analgesic activity. We are only at the beginning of understanding the mechnisms 5 behind different pain syndromes like cancer pain (e.g. tumor-induced bone cancer pain), chemotherapy-induced pain or nucleoside-induced pain which seem all to have various molecular origins. Antidepressents, anticonvulsants or opioids which describe groups of compounds used in pain treatment do not have a common pattern regarding their efficacy in treatment of pain 10 syndromes. This makes it difficult to predict the activity of new compounds in the various pain syndromes and demands a detailed characterization in multiple pain animal models. Neuropathic pain after injury or dysfunction to the peripheral or central nervous system remains a difficult clinical problem for which effective treatments are lacking (Bennett, 1994; Murphy and Reid, 2001). Anticonvulsants are used for the management of some forms of neuropathic pain (Sindrup and Jenssen, 1999; Jensen, 2002). SPM 927 (R-2-acetamido N-benzyl-3-methoxypropionamide) also called harkoseride or ADD 234037 is a novel anticonvulsant drug. It belongs to a series of functionalized amino acids which have been synthesised as a new class of anti-convulsant agents (Kohn et al. 1991). The present studies show analgesic effects of SPM 927 in rat models of cancer pain, in particular bone cancer pain, of chemotherapy-induced pain and of nucleoside analogue-induced pain. Bone is the third most common site of metastasis after lung and liver, and is 15 the primary site of metastatic disease in patients with breast, prostate and lung cancer. The bone lesions that result from metastatic disease also WO 2006/021412 PCT/EP2005/009067 -5 cause severe bone pain, which is a major clinical problem in cancer patients. This type of pain is difficult to treat due to its intermittent, progressive nature, and its aggravation by movement. The predominant symptom in this model of pain is mechanical allodynia. Thermal hyperalgesia and mechanical 5 hyperalgesia has also been demonstrated as measured by the weight bearing difference in the two hindlimbs (Medhurst et al., 2002). Treatment of bone pain in human patients is largely limited to the use of opioids, however the efficacy of potent opioids is minimal, and effective doses produce a range of debilitating side effects. Consequently, there is a clinical need for 10 new therapies that can be used to prevent, treat and alleviate tumor-induced bone pain. Candidate therapies for treatment of tumor-induced bone pain can be evaluated using a rat model as the rat is superior for testing behavioral responses to pain stimuli. One model involves the injection of rat mammary gland carcinoma cells into the marrow space of the proximal tibia 15 using an endpoint of pain assessment (Medhurst et al., 2002), which was performed on Days 7 to 15 following tumor implantation. Chemotherapy-induced pain is a form of neuropathic pain associated with neurotoxic drugs such as vinca alkaloids, e.g. vincristine and is 20 characterized by painful paresthesias and dysesthesias. The clinical antineoplastic efficacy of vincristine is limited by the development of a mixed sensorimotor neuropathy (Casey et al., 1973, Tanner et al., 1998 et al. 1998) that appears to occur in two major stages (Weiss et al., 1974). In the early stage, peripheral axons are damaged by vincristine and the principal 25 symptoms are paresthesias and dysesthesias. In the later stage, which occurs more frequently when higher doses are given for longer periods of time, axons are lost and the principal clinical finding is loss of motor function. The described vincristine rat model seems to reflect the early stage of vincristine-induced chemotherapeutic neuropathy. Whilst the underlying 30 mechanism is not fully understood as yet, it has been described to cause a disorganization of the axonal microtubule cytoskeleton, as well as an increase in the caliber of unmyelinated sensory axons (Quasthoff et al., 2002). These results demonstrated that changes in microtubule structure in WO 2006/021412 PCT/EP2005/009067 -6 nociceptive sensory neurons accompany vincristine-induced hyperalgesia. Painful peripheral neuropathy, induced by nucleoside analogues is becoming recognized as an important source of morbidity in human 5 immunodeficiency virus (HIV) infected individuals (Cohen, 2002). This severely debilitating side-effect may force abbreviation or even discontinuation of AIDS (acquired immunodeficiency syndrome) therapy (Yatvin et al., 1999). This neuropathy is characterized by a sudden onset of intense burning discomfort in both feet sparing the hands at about the 10th 10 week of treatment, which reached severe intensity over a period of days (Dubinsky et al., 1989). The biochemical mechanism underlying this neuropathic remains to be clearly established, although mitochondrial toxicity has been reported to contribute to the development of this neuropathy. Recently, it has been reported that intoxication of rats with anti 1s retroviral nucleoside analogue AIDS therapy drugs (ddC (2',3'-dideoxycytidine), ddI (2',3'-dideoxyinosine) or d4T (2',3'-didehydro 3'-deoxythymidine)), produces enhanced nociception in the rat (Joseph et al., 2004). The mechanism involved appears different from that found to contribute in other models of metabolic or toxic painful peripheral 20 neuropathy, as anti-hyperalgesic drugs effective in these models. Inhibitors of protein kinase A, protein kinase C, protein kinase G, p42/p44-mitogen activated protein kinase (ERK1/2) and nitric oxide synthase have no effect on peripheral neuropathies, and had no effect on nucleoside reverse transcriptase inhibitor-induced hypersensitivity. Intracellular calcium 25 modulators (TMB-8 and Quin-2) are the only agents capable of reversing this hypersentivity of intoxicated animals strongly suggests the role of intracellular calcium in this type of neuropathic pain. Chemotherapy, e.g. treatment with vinca alkaloids like vincristine or with 30 taxol, suramin, cisplatin, carboplatin or oxaliplatin is used for the treatment of cancer and HIV patients. Additionally, HIV or/and tumor patients are also treated with antiretrovirals or antivirals.
WO 2006/021412 PCT/EP2005/009067 -7 The use of compounds of Formula (lb) or/and Formula (11b) for treatment of tumor pain, in particular bone cancer pain, for treating chemotherapy induced pain and for treating nucleoside-induced pain, has not been reported. Thus, the present invention concerns the use of said compounds 5 of Formulae (1b) or/and (lib) for the preparation of a pharmaceutical composition for the prevention, alleviation or/and treatment of tumor pain, in particular tumor pain associated withs AIDS, bone cancer pain, pain produced during tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves or/and metastasis-induced pain such as, but 10 not limited to, metastasis-induced bone cancer pain, pain induced by metastatic disease in patients with breast, prostate or lung cancer. The present invention further concerns the use of the compounds of Formula (lb) or/and (1lb) for the preparation of a pharmaceutical composition for the prevention, alleviation or/and treatment of chemotherapy-induced pain, such 15 as, but not limited to, chemotherapy-induced neuropathic pain, vinca alkaloid-induced pain, vincristine-induced pain or/and pain induced by taxol, suramin, cisplatin, carboplatin or/and oxaliplatin. The present invention further concerns the use of the compounds of Formula (lb) or/and (11b) for the preparation of a pharmaceutical composition for the prevention, 20 alleviation or/and treatment of nucleoside- or/and nucleoside analogue induced pain, such as, but not limited to, painful peripheral neuropathy induced by nucleosides or/and nucleoside analogues, pain induced by anti tumor or/and anti-viral nucleoside analogues or/and pain induced by anti retroviral nucleoside analogues, such as AZT (3'-azidothymidine), ddC, ddl 25 or/and d4T, e.g. in AIDS therapy. The invention also concerns the use of the compounds of Formula (lb) or/and (lIb) for the preparation of a pharmaceutical composition for the prevention, alleviation or/and treatment of tumor pain, chemotherapy 30 induced pain, or/and pain induced by at least one nucleoside or/and at least one nucleoside analogue.
WO 2006/021412 PCT/EP2005/009067 Surprisingly, the application of compounds (lb) or/and (1lb), particularly (R) 2-acetamide-N-benzyl-3-methoxypropionamide (SPM 927) reduced mechanical and thermal hyperalgesia as well as mechanical and thermal allodynia in a tumor-induced bone cancer pain model, in a chemotherapy 5 induced and a nucleoside analogue-induced neuropathic pain model. A compound according to the invention has the general Formula (lb) R2 10 R NH-[-C-CNH-]n-C- R 1 0o o
R
3 15 Formula (lb) wherein 20 R is hydrogen, lower alkyl, lower alkenyl, lower alkynyl, aryl, aryl lower alkyl, heterocyclic, heterocyclic lower alkyl, lower alkyl heterocyclic, lower cycloalkyl or lower cycloalkyl lower alkyl, and R is unsubstituted or is substituted with at least one electron withdrawing group, and/or at least one electron donating group; 25 R, is hydrogen or lower alkyl, lower alkenyl, lower alkynyl, aryl lower alkyl, aryl, heterocyclic lower alkyl, lower alkyl heterocyclic, heterocyclic, lower cycloalkyl, lower cycloalkyl lower alkyl, each unsubstituted or substituted with at least one electron donating group and/or at least one electron withdrawing 30 group; and
R
2 and R 3 are independently hydrogen, lower alkyl, lower alkenyl, lower WO 2006/021412 PCT/EP2005/009067 -9 alkynyl, aryl lower alkyl, aryl, halo, heterocyclic, heterocyclic lower alkyl, lower alkyl heterocyclic, lower cycloalkyl, lower cycloalkyl lower alkyl, or Z-Y wherein R 2 and R 3 may be unsubstituted or substituted with at least one electron withdrawing group and/or at least one electron donating group; 5 Z is 0, S, S(O), NR 4 , NR' 6 , PR 4 or a chemical bond; Y is hydrogen, lower alkyl, aryl, aryl lower alkyl, lower alkenyl, lower alkynyl, halo, heterocyclic, heterocyclic lower alkyl, lower alkyl heterocyclic and Y 10 may be unsubstituted or substituted with at least one electron donating group and/or at least one electron withdrawing group, provided that when Y is halo, Z is a chemical bond, or ZY taken together is NR4NR 5
R
7 , NR4OR 5 , ONR 4
R
7 , OPR 4
R
5 , PR 4
OR
5 , 15 SNR4R 7 , NR4SR 7 , SPR4R 5 , PR4SR 7 , NR4PRR6, PR4NRsR 7 or N*R 5
R
6
R
7 , NR4C-R 5 , SCRs, NR4C-OR 5 , SC-OR 5 , NR4NR 5
-C-OR
6 ; 20 0 0 0 0 0
R'
6 is hydrogen, lower alkyl, lower alkenyl, or lower alkenyl which may be unsubstituted or substituted with at least one electron withdrawing group or/and at least one electron donating group; 25
R
4 , R 5 and R 6 are independently hydrogen, lower alkyl, aryl, aryl lower alkyl, lower alkenyl, or lower alkynyl, wherein R 4 , R 5 and R6 may independently be unsubstituted or substituted with at least one electron withdrawing group or/and at least one electron donating group; 30
R
7 is R 6 or COOR 8 or CORB, which R 7 may be unsubstituted or substituted with at least one electron withdrawing group or/and at least one electron donating group; WO 2006/021412 PCT/EP2005/009067 -10
R
8 is hydrogen or lower alkyl, or aryl lower alkyl, and the aryl or alkyl group may be unsubstituted or substituted with at least one electron withdrawing group or/and at least one electron donating group; and 5 n is 1-4; and a is 1-3. Preferably the compound according has the general Formula (lib) 10 H H H I . I I Ar-CHr-N-C-C-N-C-R 1 O R 3 0 15 Formula (lib) wherein 20 Ar is aryl, especially phenyl, which is unsubstituted or substituted with at least one halo; R 3 is -CH 2 -Q, wherein Q is lower alkoxy; and R 1 is lower alkyl, especially methyl. The present invention is also directed to a pharmaceutical composition 25 comprising a compound according to Formula (Ib) or/and Formula (lib) useful for the prevention, alleviation or/and treatment of tumor pain, in particular tumor pain associated with AIDS, bone cancer pain, pain produced during tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves or/and metastasis-induced pain such as, but 30 not limited to, metastasis-induced bone cancer pain, pain induced by metastatic disease in patients with breast, prostate or lung cancer. The present invention further concerns a pharmaceutical composition comprising a compound according to Formula (lb) or/and Formula (l1b) useful for the prevention, alleviation or/and treatment of chemotherapy-induced pain, such 35 as, but not limited to, chemotherapy-induced neuropathic pain, vinca alkaloid-induced pain, vincristine-induced pain or/and pain induced by taxol, WO 2006/021412 PCT/EP2005/009067 - 11 suramin, cisplatin, carboplatin or/and oxaliplatin. The present invention further concerns a pharmaceutical composition comprising a compound according to Formula (lb) or/and Formula (lIb) useful for the prevention, alleviation or/and treatment of nucleoside- or/and nucleoside analogue 5 induced pain, such as, but not limited to, painful peripheral neuropathy induced by nucleosides or/and nucleoside analogues, pain induced by anti tumor or/and anti-viral nucleoside analogues or/and pain induced by anti retroviral nucleoside analogues, such as AZT, ddC, ddl or/and d4T, e.g. in AIDS therapy. 10 The invention also concerns a pharmaceutical composition comprising a compound according to Formula (lb) or/and Formula (l1b) useful for the prevention, alleviation or/and treatment of tumor pain, chemotherapy induced pain, or/and pain induced by at least one nucleoside or/and at least 15 one nucleoside analogue. The "lower alkyl" groups when used alone or in combination with other groups, are lower alkyl containing from 1 to 6 carbon atoms, especially 1 to 3 carbon atoms, and may be straight chain or branched. These groups include 20 methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, amyl, hexyl, and the like. The "lower alkoxy" groups are lower alkoxy containing from 1 to 6 carbon atoms, especially 1 to 3 carbon atoms, and may be straight chain or 25 branched. These groups include methoxy, ethoxy, propoxy, butoxy, isobutoxy, tert-butoxy, pentoxy, hexoxy and the like. The "aryl lower alkyl" groups include, for example, benzyl, phenylethyl, phenylpropyl, phenylisopropyl, phenylbutyl, diphenylmethyl, 1,1 30 diphenylethyl, 1,2-diphenylethyl, and the like. The term "aryl", when used alone or in combination, refers to an aromatic group which contains from 6 up to 18 ring carbon atoms and up to a total of WO 2006/021412 PCT/EP2005/009067 - 12 25 carbon atoms and includes the polynuclear aromatics. These aryl groups may be monocyclic, bicyclic, tricyclic or polycyclic and are fused rings. A polynuclear aromatic compound as used herein, is meant to encompass bicyclic and tricyclic fused aromatic ring systems containing from 10-18 ring 5 carbon atoms and up to a total of 25 carbon atoms. The aryl group includes phenyl, and the polynuclear aromatics e.g., naphthyl, anthracenyl, phenanthrenyl, azufenyl and the like. The aryl group also includes groups like ferrocenyl. Aryl groups may be unsubstituted or mono or polysubstituted with electron withdrawing or/and electron donating groups as described 10 below. "Lower alkenyl" is an alkenyl group containing from 2 to 6 carbon atoms and at least one double bond. These groups may be straight chained or branched and may be in the Z or E form. Such groups include vinyl, 15 propenyl, 1-butenyl, isobutenyl, 2-butenyl, 1-pentenyl, (Z)-2-pentenyl, (E)-2 pentenyl, (Z)-4-methyl-2-pentenyl, (E)-4-methyl-2-pentenyl, pentadienyl, e.g., 1, 3 or 2,4-pentadienyl, and the like. The term "lower alkynyl" is an alkynyl group containing 2 to 6 carbon atoms 20 and may be straight chained as well as branched. It includes such groups as ethynyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1 pentynyl, 3-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl and the like. The term "lower cycloalkyl" when used alone or in combination is a 25 cycloalkyl group containing from 3 to 18 ring carbon atoms and up to a total of 25 carbon atoms. The cycloalkyl groups may be monocyclic, bicyclic, tricyclic, or polycyclic and the rings are fused. The cycloalkyl may be completely saturated or partially saturated. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, 30 cyclohexenyl, cyclopentenyl, cyclooctenyl, cycloheptenyl, decalinyl, hydroindanyl, indanyl, fenchyl, pinenyl, adamantyl, and the like. Cycloalkyl includes the cis or trans forms. Cycloalkyl groups may be unsubstituted or mono or polysubstituted with electron withdrawing or/and electron donating WO 2006/021412 PCT/EP2005/009067 -13 groups as described below. Furthermore, the substituents may either be in endo or exo positions in the bridged bicyclic systems. The term "electron-withdrawing and electron donating" refer to the ability of a 5 substituent to withdraw or donate electrons, respectively, relative to that of hydrogen if the hydrogen atom occupied the same position in the molecule. These terms are well understood by one skilled in the art and are discussed in Advanced Organic Chemistry, by J. March, John Wiley and Sons, New York, NY, pp.16-18 (1985) and the discussion therein is incorporated herein 10 by reference. Electron withdrawing groups include halo, including bromo, fluoro, chioro, iodo and the like; nitro, carboxy, lower alkenyl, lower alkynyl, formyl, carboxyamido, aryl, quaternary ammonium, haloalkyl such as trifluoromethyl, aryl lower alkanoyl, carbalkoxy and the like. Electron donating groups include such groups as hydroxy, lower alkoxy, including 15 methoxy, ethoxy and the like; lower alkyl, such as methyl, ethyl, and the like; amino, lower alkylamino, di(loweralkyl) amino, aryloxy such as phenoxy, mercapto, lower alkylthio, lower alkylmercapto, disulfide (lower alkyldithio) and the like. One of ordinary skill in the art will appreciate that some of the aforesaid substituents may be considered to be electron donating or electron 20 withdrawing under different chemical conditions. Moreover, the present invention contemplates any combination of substituents selected from the above-identified groups. The term "halo" includes fluoro, chloro, bromo, iodo and the like. 25 The term "acyl" includes lower alkanoyl containing from 1 to 6 carbon atoms and may be straight chains or branched. These groups include, for example, formyl, acetyl, propionyl, butyryl, isobutyryl, tertiary butyryl, pentanoyl and hexanoyl. 30 As employed herein, a heterocyclic group contains at least one sulfur, nitrogen or oxygen ring atom, but also may include several of said atoms in the ring. The heterocyclic groups contemplated by the present invention WO 2006/021412 PCT/EP2005/009067 - 14 include heteroaromatics and saturated and partially saturated heterocyclic compounds. These heterocyclics may be monocyclic, bicyclic, tricyclic or polycyclic and are fused rings. They may preferably contain up to 18 ring atoms and up to a total of 17 ring carbon atoms and a total of up to 25 5 carbon atoms. The heterocyclics are also intended to include the so-called benzoheterocyclics. Representative heterocyclics include furyl, thienyl, pyrazolyl, pyrrolyl, methylpyrroly, imidazolyl, indolyl, thiazolyl, oxazolyl, isothiazolyl, isoxazolyl, piperidyl, pyrrolinyl, piperazinyl, quinolyl, triazolyl, tetrazolyl, isoquinolyl, benzofuryl, benzothienyl, morpholinyl, benzoxazolyl, 10 tetrahydrofuryl, pyranyl, indazolyl, purinyl, indolinyl, pyrazolindinyl, imidazolinyl, imadazolindinyl, pyrrolidinyl, furazanyl, N-methylindolyl, methylfuryl, pyridazinyl, pyrimidinyl, pyrazinyl, pyridyl, epoxy, aziridino, oxetanyl, azetidinyl, the N-oxides of the nitrogen containing heterocycles, such as the N-oxides of pyridyl, pyrazinyl, and pyrimidinyl and the like. 15 Heterocyclic groups may be unsubstituted or mono or polysubstituted with electron withdrawing or/and electron donating groups. The preferred heterocyclics are thienyl, furyl, pyrrolyl, benzofuryl, benzothienyl, indolyl, methylpyrrolyl, morpholinyl, pyridiyl, pyrazinyl, 20 imidazolyl, pyrimidinyl, or pyridazinyl. The preferred heterocyclic is a 5 or 6 membered heterocyclic compound. The especially preferred heterocyclic is furyl, pyridyl, pyrazinyl, imidazolyl, pyrimidinyl, or pyridazinyl. The most preferred heterocyclics are furyl and pyridyl. 25 The preferred compounds are those wherein n is 1, but di (n=2), tri (n=3) and tetrapeptides (n=4) are also contemplated to be within the scope of the invention. The preferred values of R is aryl lower alkyl, especially benzyl especially 30 those wherein the phenyl ring thereof is unsubstituted or substituted with electron donating groups or/and electron withdrawing groups, such as halo (e.g., F).
WO 2006/021412 PCT/EP2005/009067 -15 The preferred R 1 is H or lower alkyl. The most preferred R 1 group is methyl. The preferred electron donating substituents or/and electron withdrawing substituents are halo, nitro, alkanoyl, formyl, arylalkanoyl, aryloyl, carboxyl, 5 carbalkoxy, carboxamido, cyano, sulfonyl, sulfoxide, heterocyclic, guanidine, quaternary ammonium, lower alkenyl, lower alkynyl, sulfonium salts, hydroxy, lower alkoxy, lower alkyl, amino, lower alkylamino, di(loweralkyl) amino, amino lower alkyl, mercapto, mercaptoalkyl, alkylthio, and alkyldithio. The term "sulfide" encompasses mercapto, mercapto alkyl and alkylthio, 10 while the term disulfide encompasses alkyldithio. Especially preferred electron donating or/and electron withdrawing groups are halo or lower alkoxy, most preferred are fluoro or methoxy. These preferred substituents may be present on any one of the groups in Formula (Ib) or/and (11b), e.g. R,
R
1 , R2, R 3 , R 4 , R 5 , R 6 , R' 6 , R 7 , R 8 and/or R 5 o as defined herein. 15 The ZY groups representative of R 2 and R 3 include hydroxy, alkoxy, such as methoxy, ethoxy, aryloxy, such as phenoxy; thioalkoxy, such as thiomethoxy, thioethoxy; thioaryloxy such as thiophenoxy; amino; alkylamino, such as methylamino, ethylamino; arylamino, such as anilino; lower dialkylamino, 20 such as, dimethylamino; trialkyl ammonium salt, hydrazino; alkylhydrazino and arylhydrazino, such as N-methylhydrazino, N-phenylhydrazino, carbalkoxy hydrazine, aralkoxycarbonyl hydrazino, aryloxycarbony hydrazino, hydroxylamino, such as N-hydroxylamino (-NH-OH), lower alkoxy amino [(NHOR 1 8 ) wherein R 18 is lower alkyl], N-lower alkylhydroxyl amino 25 [(NR 1 )OH wherein R 18 is lower alkyl], N-lower alkyl-O-lower alkylhydroxyamino, i.e., [N(R 18
)OR
1 wherein R 1 8 and R 1 are independently lower alkyl], and o-hydroxylamino (-0-NH 2 ); alkylamido such as acetamido; trifluoroacetamido; lower alkoxyamino, (e.g., NH(OCH 3 ); and heterocyclicamino, such as pyrazoylamino. 30 The preferred heterocyclic groups representative of R 2 and R 3 are monocyclic 5- or 6-membered heterocyclic moieties of the formula: WO 2006/021412 PCT/EP2005/009067 - 16 A 5 E
R
5 o G L (CH), . 10 or those corresponding partially or fully saturated form thereof wherein n is 0 or 1; and
R
50 is H or an electron withdrawing group or electron donating group; 15 A, E, L, J and G are independently CH, or a heteroatom selected from the group consisting of N, 0, S; but when n is 0, G is CH, or a heteroatom selected from the group consisting of NH, 0 and S with the proviso that at most two of A, E, L, J and G are 20 heteroatoms. When n is 0, the above heteroaromatic moiety is a five membered ring, while if n is 1, the heterocyclic moiety is a six membered monocyclic heterocyclic moiety. The preferred heterocyclic moieties are those aforementioned 25 heterocyclics which are monocyclic. If the ring depicted hereinabove contains a nitrogen ring atom, then the N oxide forms are also contemplated to be within the scope of the invention. 30 When R 2 or R 3 is a heterocyclic of the above formula, it may be bonded to the main chain by a ring carbon atom. When n is 0, R 2 or R 3 may additionally be bonded to the main chain by a nitrogen ring atom.
WO 2006/021412 PCT/EP2005/009067 - 17 Other preferred moieties of R 2 and R 3 are hydrogen, aryl, e.g., phenyl, aryl alkyl, e.g., benzyl and alkyl. It is to be understood that the preferred groups of R 2 and R 3 may be 5 unsubstituted or mono or poly substituted with electron donating or/and electron withdrawing groups. It is preferred that R 2 and R 3 are independently hydrogen, lower alkyl, which is either unsubstituted or substituted with electron withdrawing groups or/and electron donating groups, such as lower alkoxy (e.g., methoxy, ethoxy, and the like), N-hydroxylamino, N-lower 10 alkylhydroxyamino, N-loweralkyl-O-loweralkyl and alkylhydroxyamino. It is preferred that one of R 2 and R 3 is hydrogen. It is preferred that n is one. 15 It is more prefered that n=1 and one of R 2 and R 3 is hydrogen. It is especially preferred that in this embodiment, R 2 is hydrogen and R 3 is lower alkyl or ZY; Z is 0, NR 4 or PR 4 ; Y is hydrogen or lower alkyl; ZY is NR 4 NRsR 7 , NR 4
OR
5 ,
ONR
4
R
7 , NR 4
C-R
5 or NR 4
C-OR
5 . 20 || || 0 0 In another especially preferred embodiment, n=1, R 2 is hydrogen and R 3 is lower alkyl which may be substituted or unsubstituted with an electron 25 donating or electron withdrawing group, NR 4
OR
5 , or ONR 4
R
7 , In yet another especially preferred embodiment, n = 1, R 2 is hydrogen and
R
3 is lower alkyl which is unsubstituted or substituted with hydroxy or loweralkoxy, NR 4
OR
5 or ONR 4 R, wherein R 4 , R 5 and R 7 are independently 30 hydrogen or lower alkyl, R is aryl lower alkyl, which aryl group may be unsubstituted or substituted with an electron withdrawing group and R 1 is lower alkyl. In this embodiment it is most preferred that aryl is phenyl, which is unsubstituted or substituted with halo.
WO 2006/021412 PCT/EP2005/009067 - 18 It is preferred that R 2 is hydrogen and R 3 is hydrogen, an alkyl group which is unsubstituted or substituted by at least an electron donating or electron withdrawing group or ZY. In this preferred embodiment, it is more preferred that R 3 is hydrogen, an alkyl group such as methyl, which is unsubstituted or 5 substituted by an electron donating group, or NR 4
OR
5 or ONR 4
R
7 , wherein
R
4 , R 5 and R 7 are independently hydrogen or lower alkyl. It is preferred that the electron donating group is lower alkoxy, and especially methoxy or ethoxy. 10 It is preferred that R 2 and R 3 are independently hydrogen, lower alkyl, or ZY; Z is 0, NR 4 or PR 4 ; Y is hydrogen or lower alkyl or ZY is NR 4 RsR 7 , NR 4
OR
5 , ONR 4
R
7 , NR 4
C-R
5 or NR 4 C-ORs. 15 11 11 0 0 It is also preferred that R is aryl lower alkyl. The most preferred aryl for R is phenyl. The most preferred R group is benzyl. In a preferred embodiment, the aryl group may be unsubstituted or substituted with an electron donating 20 or electron withdrawing group. If the aryl ring in R is substituted, it is most preferred that it is substituted with an electron withdrawing group, especially on the aryl ring. The most preferred electron withdrawing group for R is halo, especially fluoro. 25 The preferred R 1 is lower alkyl, especially methyl. It is more preferred that R is aryl lower alkyl and R 1 is lower alkyl. Further preferred compounds are compounds of Formula (lb) wherein n is 1; 30 R 2 is hydrogen; R 3 is hydrogen, a lower alkyl group, especially methyl which is substituted by an electron donating or electron withdrawing group or ZY; R is aryl, aryl lower alkyl, such as benzyl, wherein the aryl group is unsubstituted or substituted with an electron donating or electron withdrawing group and R 1 is lower alkyl. In this embodiment, it is more WO 2006/021412 PCT/EP2005/009067 -19 preferred that R 3 is hydrogen, a lower alkyl group, especially methyl, which may be substituted by electron donating group, such as lower alkoxy, (e.g., methoxy, ethoxy and the like), NR 4 0R 5 or ONR 4
R
7 wherein these groups are defined hereinabove. 5 The most preferred compounds utilized are those of the Formula (Ilb): H H H 10 | | 1 Ar-CH 2
-N-C-C-N-C-R
1 O R 3 0 15 Formula (lib) wherein Ar is aryl, especially phenyl, which is unsubstituted or substituted with at least one electron donating group or electron withdrawing group, especially 20 halo, R, is lower alkyl, especially containing 1-3 carbon atoms; and
R
3 is as defined herein, but especially hydrogen, loweralkyl, which is 25 unsubstituted or substituted by at least an electron donating group or electron withdrawing group or ZY. It is even more preferred that R 3 is, in this embodiment, hydrogen, an alkyl group which is unsubstituted or substituted by an electron donating group, NR 4 OR or ONR 4
R
7 . It is most preferred that
R
3 is CH 2 -Q, wherein Q is lower alkoxy, especially containing 1-3 carbon 30 atoms; NR 4
OR
5 or ONR 4
R
7 wherein R 4 is hydrogen or alkyl containing 1-3 carbon atoms, R 5 is hydrogen or aikyl containing 1-3 carbon atoms, and R 7 is hydrogen or alkyl containing 1-3 carbon atoms. The most preferred R 1 is CH 3 . The most preferred R 3 is CH 2 -Q, wherein Q is 35 methoxy.
WO 2006/021412 PCT/EP2005/009067 -20 The most preferred aryl is phenyl. The most preferred halo is fluoro. The most preferred compounds include: (R)-2-acetamido-N-benzyl-3-methoxy-propionamide; 5 O-methyl-N-acetyl-D-serine-m-fluorobenzyl-amide; O-methyl-N-acetyl-D-serine-p-fluorobenzyl-amide; N-acetyl-D-phenylglycine benzylamide; D-1,2-(N,O-dimethylhydroxylamino)-2-acetamide acetic acid benzylamide; D-1,2-(O-methylhydroxylamino)-2-acetamido acetic acid benzylamide. 10 It is to be understood that the various combinations and permutations of the Markush groups of R 1 , R 2 , R 3 , R and n described herein are contemplated to be within the scope of the present invention. Moreover, the present invention also encompasses compounds and compositions which contain one or more 15 elements of each of the Markush groupings in R 1 , R 2 , R 3 , n and R and the various combinations thereof. Thus, for example, the present invention contemplates that R 1 may be one or more of the substituents listed hereinabove in combination with any and all of the substituents of R 2 , R 3 , and R with respect to each value of n. 20 The compounds utilized in the present invention may contain one or more asymmetric carbons and may exist in racemic and optically active forms. The configuration around each asymmetric carbon can be either the D or L form. It is well known in the art that the configuration around a chiral carbon 25 atoms can also be described as R or S in the Cahn-Prelog-Ingold nomenclature system. All of the various configurations around each asymmetric carbon, including the various enantiomers and diastereomers as well as racemic mixtures and mixtures of enantiomers, diastereomers or both are contemplated by the present invention. 30 In the principal chain, there exists asymmetry at the carbon atom to which the groups R 2 and R 3 are attached. When n is 1, the compounds of the present invention is of the formula WO 2006/021412 PCT/EP2005/009067 -21
R
2 0 1 H || 5 R-NH-C-C-N-C--R 1 (Ill) I1 I O R 3 wherein R, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R' 6 , R 7 , R 8 , R 6 o, Z and Y are as defined 10 previously. As used herein, the term configuration shall refer to the configuration around the carbon atom to which R 2 and R 3 are attached, even though other chiral centers may be present in the molecule. Therefore, when referring to a 15 particular configuration, such as D or L, it is to be understood to mean the D or L stereoisomer at the carbon atom to which R 2 and R 3 are attached. However, it also includes all possible enantiomers and diastereomers at other chiral centers, if any, present in the compound. 20 The compounds of the present invention are directed to all the optical isomers, i.e., the compounds of the present invention are either the L stereoisomer or the D-stereoisomer (at the carbon atom to which R 2 and R 3 are attached). These stereoisomers may be found in mixtures of the L and D stereoisomer, e.g., racemic mixtures. The D stereoisomer is preferred. 25 More preferred is a compound of Formula (ill) in the R configuration, preferably substantially enantiopure, wherein the substituent R is benzyl which is unsubstituted or substituted with at least one halo group, wherein R 3 is CH 2 -Q, wherein Q is lower alkoxy containing 1-3 carbon atoms and 30 wherein R 1 is methyl. Preferably R is unsubstituted benzyl or benzyl substituted with at least one halo group which is a fluoro group. Depending upon the substituents, the present compounds may form addition salts as well. All of these forms are contemplated to be within the scope of 35 this invention including mixtures of the stereoisomeric forms.
-22 The manufacture of the utilized compounds Is described In U.S. Patent Nos. 5,378,729 and 5,773.475, the contents of both of which are Incorporated by reference. The compounds utilized In the present invention are useful as such as depicted in the Formulae (ib) or/and (lib) or can be employed In the form of salts In view of Its basic. nature by the presence of the free amino group. Thus, the compounds of Formulae (Ib) or/and (lib) form salts with a wide variety of acids, inorganic and organc, including pharmaceutically acceptable acids. The salts with therapeutically acceptable acids are of course useful in the preparation of formulation where enhanced water solubility is most advantageous. These pharmaceutically acceptable salts have also therapeutic efficacy. These salts Include salts of Inorganic acids such as hydrochloric, hydrolodic, hydrobromic, phosphoric, metaphosphoric, nitic acid and sulfuric acids as well as salts of organic acids, such as tartaric, acetic, citric, malic, benzoic, perchloric, glycolic, gluconlc, succinic, aryl sulfonic, (e.g., p-toluene sulfonic adds, benzenesufonic), phosphoric, malonic, and the like. The present invention is further directed to a pharmaceutical composition comprising: (a) a compound of formula (Ib) or/and (1lb), and (b) a further active agent for the prevention, alleviation or/and treatment of a viral infection such as HIV infection including AIDS, of cancer, such as breast cancer, prostate cancer, lung cancer, bone cancer, metastatic disease or/and of tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves, wherein the pharmaceutical composition is a single dose form.
C:WRPortbl\DCC\XT329K646.I DOC-I 111112010 -22A The present Invention is further directed to a method for the prevention, alleviation or/and treatment of a disease or condition as described above in a mammal, Including a human being, comprising administering at least one compound of formulae (Ib) orland (lib). It Is preferred that the compound utilized In the present invention Is used In therapeutically effective amounts. The physician will determine the dosage of the present therapeutic agents which will be most suitable and it will vary with the form of administration and the particular compound chosen, and furthermore, it will vary with the patient under treatment, the age of the patient, the type of malady being treated. He WO 2006/021412 PCT/EP2005/009067 - 23 will generally wish to initiate treatment with small dosages substantially less than the optimum dose of the compound and increase the dosage by small increments until the optimum effect under the circumstances is reached. When the composition is administered orally, larger quantities of the active 5 agent will be required to produce the same effect as a smaller quantity given parenterally. The compounds are useful in the same manner as comparable therapeutic agents and the dosage level is of the same order of magnitude as is generally employed with these other therapeutic agents. 10 In a preferred embodiment, -the compounds of the present invention are administered in amounts ranging from about 1 mg to about 100 mg per kilogram of body weight per day, more preferably in amounts ranging from about 1 mg to about 10 mg per kilogram of body weight per day. This dosage regimen may be adjusted by the physician to provide the optimum 15 therapeutic response. Patients in need thereof may be treated with doses of the compound of the present invention of at least 50 mg/day, preferably of at least 200 mg/day, more preferably of at least 300 mg/day and most preferably of at least 400 mg/day. Generally, a patient in need thereof may be treated with doses at a maximum of 6 g/day, more preferably a maximum 20 of I g/day and most preferably a maximum of 600 mg/day. In some cases, however, higher or lower doses may be needed. In another preferred embodiment, the daily doses are increased until a predetermined daily dose is reached which is maintained during the further 25 treatment. In yet another preferred embodiment, several divided doses may be administered daily. For example, three doses per day may be administered, preferably two doses per day. It is more preferred to administer a single 30 dose per day. In yet another preferred embodiment, an amount of the compounds of the present invention may be administered which results in a plasma WO 2006/021412 PCT/EP2005/009067 -24 concentration of 0.1 to 15 pg/ml (trough) and 5 to 18.5 pg/mi (peak), calculated as an average over a plurality of treated subjects. The compounds of Formulae (Ib) or/and (Ilb) may be administered in a 5 convenient manner, such as by oral, intravenous (where water soluble), intramuscular, intrathecal or subcutaneous routes. Oral or/and Lv. administration is preferred. The pharmaceutical composition of the present invention may be prepared 10 for the treatment regimen as described above, in particular for the treatment with doses as described above, to effect plasma concentrations as described above, for administration periods or/and administration routes as specified in the embodiments of the present invention as described above. 1s In another preferred embodiment, the method of the present invention as described above for the treatment of a mammal including a human being in need thereof comprises administering a compound of the present invention in combination with administering a further active agent for the prevention, alleviation or/and treatment of a viral infection, such as retroviral infection, 20 HIV infection including AIDS, of cancer such as breast cancer, prostate cancer, lung cancer, bone cancer, metastatic disease, or/and of tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves. The compound of the present invention and the further active agent may be administered together, i.e. in a single dose form, or may be 25 administered separately, i.e. in a separate dose form. Thus, the pharmaceutical composition of the present invention may comprise a compound of the present invention as defined above and may further comprise a further active agent for the prevention, alleviation or/and treatment of a viral infection such as retroviral infection, HIV infection 30 including AIDS, of cancer such as breast cancer, prostate cancer, lung cancer, bone cancer, metastatic disease or/and of tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves. The pharmaceutical composition may comprise a single dose form or may WO 2006/021412 PCT/EP2005/009067 -25 comprise a separate dose form comprising a first composition comprising a compound of the present invention as defined above and a second composition comprising the further active agent. 5 The compounds of the present invention may be used for the preparation of a pharmaceutical composition as described above. The compounds of Formulae (lb) or/and (lb) may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may 10 be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly into the fool of the diet. For oral therapeutic administration, the active compound of Formulae (lb) or/and (lib) may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, 15 wafers, and the like. Such compositions and preparations should contain at least 1 % of active compound of Formulae (Ib) or/and (11b). The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 5 to about 80 % of the weight of the unit. The amount of active compound of Formulae (lb) or/and (11b) in such 20 therapeutically useful compositions is such that a suitable dosage will be obtained. Preferred compositions or preparations according to the present invention contains between about 10 mg and 6 g active compound of Formulae (Ib) or/and (lIb). 25 The tablets, troches, pills, capsules and the like may also contain the following: A binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or 30 saccharin may be added or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier.
WO 2006/021412 PCT/EP2005/009067 -26 Various other materials may be present as coatings or otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as 5 preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and formulations. For example, sustained release dosage 10 forms are contemplated wherein the active ingredient is bound to an ion exchange resin which, optionally, can be coated with a diffusion barrier coating to modify the release properties of the resin. The active compound may also be administered parenterally or 15 intraperitoneally. Dispersions can also be prepared in glycerol, liquid, polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms, 20 The pharmaceutical forms suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture 25 and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be 30 maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for WO 2006/021412 PCT/EP2005/009067 -27 example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents 5 delaying absorption, for example, aluminium monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered 10 sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum 15 drying the freeze-drying technique plus any additional desired ingredient from previously sterile-filtered solution thereof. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agent, 20 isotonic and absorption delaying agents for pharmaceutical active substances as well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. 25 It is especially advantageous to formulate parenteral compositions in dosage unit form or ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a 30 predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specifics for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active WO 2006/021412 PCT/EP2005/009067 -28 material an the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such as active material for the treatment of disease in living subjects having a diseased condition in which bodily health is impaired as herein disclosed in detail. 5 The principal active ingredient is compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in dosage unit form as hereinbefore described. A unit dosage form can, for example, contain the principal active compound in 10 amounts ranging from about 10 mg to about 6 g. Expressed in proportions, the active compound is generally present in from about I to about 750 mg/ml of carrier. In the case of compositions containing supplementary active ingredients, the dosages are determined by reference to the usual dose and manner of administration of the said ingredients. 15 As used herein the term "patient" or "subject" refers to a warm blooded animal, and preferably mammals, such as, for example, cats, dogs, horses, cows, pigs, mice, rats and primates, including humans. The preferred patient is a human. 20 The term "treat" refers to either relieving the pain associated with a disease or condition, to curing or alleviating the patient's disease or condition. The compounds of the present invention are administered to a patient 25 suffering from the aforementioned type of disorder in an effective amount. These amounts are equivalent to the therapeutically effective amounts described hereinabove. The following example shows the properties of SPM 927 in reducing so mechanical and thermal hyperalgesia as well as mechanical and thermal allodynia in a tumor-induced bone cancer pain model, in a chemotherapy induced and a nucleoside analogue-induced neuropathic pain model.
WO 2006/021412 PCT/EP2005/009067 -29 The used substance was SPM 927 which is the synonym for Harkoseride. The standard chemical nomenclature is (R)-2-acetamide-N-benzyl-3 methoxypropionamide. 5 Figure Legend Figure 1 describes testing of mechanical allodynia in a bone cancer pain model (rats). Bone cancer rats were treated with increasing concentrations of SPM 927 (10 mg, 20 mg and 40 mg) and compared with morphine treated 10 bone cancer rats, bone cancer rats without treatment (cells only) and control rats. Figure 2 describes testing of thermal paw stimulation at days 14 and 15 in a bone cancer pain model (rats). Bone cancer rats were treated with 15 increasing concentrations of SPM 927 (3 mg, 10 mg and 30 mg) and compared with morphine treated bone cancer rats, bone cancer rats without treatment (cells only) and control rats. Figure 3 describes testing of weight bearing differences in a bone cancer 20 pain model (rats). Bone cancer rats were treated with increasing concentrations of SPM 927 (10 mg, 20 mg and 40 mg) and compared with morphine treated bone cancer rats, bone cancer rats without treatment (cells only) and control rats. 25 Figure 4 describes the effect of increasing concentations of SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) on thermal allodynia of cold bath test in a chemotherapy-induced pain model (rats treated with vincristine), compared with morphine (3 mg/kg). 30 Figure 5 describes the effect of increasing concentations of SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) on thermal allodynia of hot plate test at 380 C in a chemotherapy-induced pain model (rats treated with vincristine), compared with morphine (3 mg/kg).
WO 2006/021412 PCT/EP2005/009067 - 30 Figure 6 describes the effect of increasing concentations of SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) on thermal hyperalgesia of hot plate test at 52*C in a chemotherapy-induced pain model (rats treated with vincristine), 5 compared with morphine (3 mg/kg). Figure 7 describes the effect of increasing concentations of SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) on mechanical hyperalgesia of paw pressure test in a chemotherapy-induced pain model (rats treated with 10 vincristine), compared with morphine (3 mg/kg). Figure 8 describes the effect of increasing concentations of SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) on mechanical allodynia of Frey hair stimulation tests in a chemotherapy-induced pain model (rats treated with 15 vincristine), compared with morphine (3 mg/kg). Figure 9 describes the effect of increasing concentations of i.p.
administered SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) in a thermal allodynia test (cold bath) in a nucleoside-induced pain model (rats treated 20 with ddC), compared with the effect of morphine (3 mg/kg administered s.c.). Figure 10 describes the effect of increasing concentations of i.p.
administered SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) in a mechanical allodynia test (brushing at D20) in a nucleoside-induced pain model (rats 25 treated with ddC), compared with the effect of morphine (3 mg/kg administered s.c.). Figure 11 describes the effect of increasing concentations of i.p.
administered SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) in a mechanical 30 allodynia test (von Frey hair) in a nucleoside-induced pain model (rats treated with ddC), compared with the effect of morphine (3 mg/kg administered s.c.).
WO 2006/021412 PCT/EP2005/009067 -31 Figure 12 describes the effect of increasing concentations of i.p.
administered SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) in a thermal hyperalgesia test (hot plate at 52*C) in a nucleoside-induced pain model (rats treated with ddC), compared with the effect of morphine (3 mg/kg 5 administered s.c.). Figure 13 describes the effect of increasing concentations of i.p.
administered SPM 927 (3 mg/kg, 10 mg/kg and 30 mg/kg) in a mechanical hyperalgesia test (paw pressure) in a nucleoside-induced pain model (rats 10 treated with ddC), compared with the effect of morphine (3 mg/kg administered s.c.). Example 15 The effect of systemic administration of SPM 927 was examined in rats in a tumor-induced bone cancer pain model, in a chemotherapy-induced and a nucleoside analogue-induced neuropathic pain model. SPM 927 reduced mechanical and thermal hyperalgesia as well as mechanical and thermal allodynia in these models. It was demonstrated that SPM 927 is useful as an 20 analgesic for treating bone cancer pain, chemotherapy- and nucleoside induced neuropathy and is overall more active than morphine. Materials and methods Bone cancer rat model Cell culture 25 Cells were cultured in medium containing RPMI-1640 (Gibco, 500ml), 10% heat-inactivated fetal bovine serum (Hyclone), L-glutamine (final concentration 2mM, from Gibco) and antibiotic solution (final concentration 10OU/ml penicillin and 100ug/ml streptomycin sulfate, from Gibco). Cells were released from the tissue culture flask by brief exposure to 0.1% trypsin 30 (Gibco) and then prepared for injection as follows: cells were centrifuged for 10 minutes at approximately 1,200 rpm. The resulting pellet was washed WO 2006/021412 PCT/EP2005/009067 - 32 twice in Phosphate Buffered Saline (PBS, Mediatech) containing no calcium or magnesium. Final pellet was re-suspended in PBS and cell count taken using a hemocytometer. Cells were diluted to achieve final concentrations for injection and kept on ice until injection was performed. 5 Surgery After one week of quarantine, either culture media or 3 X 104 syngeneic MRMT-1 rat mammary gland carcinoma cells were injected into the medullary cavity of the proximal tibia of each rat. Procedurally, the animal 10 was first anesthetized with ketamine/xylazine and the right leg area shaved and treated with an iodine solution and cleaned with a 70% ethanol solution. A 1-cm rostral-caudal incision was made in the skin over the top half of the tibia. Blunt-dissection was performed to expose the tibia, ensuring minimal damage to muscles or blood vessels. Using a 23-gauge needle, the tibia 15 was pierced 1-3mm below the knee joint. The needle was inserted at an angle to enable it to be pushed down into the intramedullary canal of the bone. Once a pathway to the intramedullary canal was opened, the 23 gauge needle was removed and replaced by a blunt needle attached to a 5 lil Hamilton syringe. A 3 gI volume of culture medium + vehicle or tumor cells 20 + vehicle was injected into the intramedullary cavity. The cancer cells were slowly injected while simultaneously removing the syringe, enabling the cells to fill the space in the cavity. Following the injection, the injection site was closed using bone wax. The wound was then closed using surgical staples. Post-operative care and observation was carried out until the animal 25 recovered consciousness Behavior measurements Dosing On day 8 or 15, the rats were dosed with a single injection of vehicle, 30 reference compound or test article 20 minutes prior to initiation of testing for mechanical allodynia and approximately 40 minutes prior to initiation of testing for thermal hyperalgesia. Based on the pharmacological activity of SPM 927, the tests needed to be performed no longer than 90 minutes after WO 2006/021412 PCT/EP2005/009067 -33 drug treatment. On day 9, the rats were dosed with reference compound or test article 20 minutes prior to initiation of testing for weight bearing. Nociceptive evaluations 5 On days 7, 8, 14 and 15, pain assessment tests were performed. On day 7, 14, the baseline evaluations were performed. All animals received an i.p. injection of saline approximately 20 minutes prior to baseline testing. On day 8, 15 starting approximately 20 minutes after the test/reference article injection, the animals underwent a series of nociceptive evaluations. The 10 order of testing remained the same for all animals. The animals were first evaluated for mechanical allodynia and then thermal hyperalgesia. The animals underwent testing for weight bearing on day 9 and 15. The animals were first evaluated for baseline weight bearing responses. Following the baseline measurements, rats were injected with the test/reference article 16 and at least 20 minutes later, the animals underwent another weight bearing analysis. Mechanical Allodynia A Von Frey test of mechanical allodynia was performed on the affected 20 (right) hind limb of all animals at baseline (day 7) and on day 8. In this test, the rats were placed in a small plexiglas box with a wire mesh floor. After approximately 10 min habituation, a series of thin nylon fibers were applied from below, through the cage floor, and pressed against the plantar surface of the hindpaw. The rats were unrestrained and un-handled during the test. 25 The diameters of the filaments provided a logarithmic scale of force exerted and thus a linear and interval scale of perceived intensity. The fiber with the weakest force was tested first and is below the normal threshold of detection for most rats. Each successively strong fiber was tested, in each case using a force required to just start to bend the monofilament. When the rat lifted its 30 paw in response to the pressure, the filament size was recorded and a weaker filament was used next. The withdrawal threshold was determined according to Chapman's "up-down" method involving the use of successively larger and smaller fibers to focus in on the withdrawal threshold. Significant WO 2006/021412 PCT/EP2005/009067 - 34 increase in allodynia was based on the comparison of group mean values. Test of Thermal Hyperalgesia The animals underwent testing for thermal hyperalgesia at baseline (day 7, 5 14) and on day 8, 15. Each rat was placed in an individual plexiglas chamber on an elevated heated glass surface for approximately 10 min to habituate. When the animal was at rest, a fiber-optic heat source was guided beneath the glass and aimed at the animal's right hindpaw. The infrared beam was turned on and when the rat lifted or moved its paw, the beam shut off 10 automatically. A timer in the machine recorded the latency to remove the foot, which was taken to signify the time for the animal to detect pain resulting from the heat. If the rat had not moved within 25 sec, the heat source automatically shut off, ensuring that there was no damage to the paw. Only the affected hindpaw was tested. This process was repeated at 15 least twice for each rat, about 3 min apart. If the latencies were within 2 sec of each other, they were averaged. If the latencies differed by more than 2 sec, the rat was tested until there were 2 latencies within 2 sec and these two numbers were averaged. Group means for latency to withdraw the paw were compared across groups with a lower latency indicating more pain 20 sensitivity. Weight Bearing After the animals in groups 4 - 6 were re-randomized to drug treatment groups, on day 9, 15 all animals underwent weight-bearing testing. Weight 25 bearing of affected hind limbs was assessed as the difference in weight borne by the ipsilateral compared to the contralateral limb. Experimentally, the rats were placed in a plexiglas chamber designed so that each hind paw is resting on a separate transducer pad that records the distribution of the animal's body weight on each paw. Five readings from each paw was 30 acquired and then averaged with results expressed as weight bearing difference (WBD; contralateral reading - ipsilateral reading). Rats were allocated eight (8) animals per group based on body weight on the day following arrival. The mean bodyweights for each group was reviewed to WO 2006/021412 PCT/EP2005/009067 - 35 ensure that the mean values and standard deviation satisfy the assumption of homogeneity. Based on the Von Frey testing results on day 8, the animals in groups 4 - 6 were re-randomized to new treatment groups for the weight bearing test to prevent any bias from their previous group assignment. 5 Animals in groups I - 3 remained within their designated groups since these were animals that were naive to the SPM 927 treatment. Vincristine-induced pain model Animal treatment 10 For this study, 86 female Dark Agouti rats (150-200 g) were used (Harlan, Gannat, France). They were group-housed (3 animals per cage) and maintained in a room with controlled temperature (21-22*C) and a reversed light-dark cycle (12h/12h) with food and water available ad libitum. All experiments were carried out in accordance with institutional guidelines. 15 Vincristine intoxication was achieved by daily injection of vincristine (0.15 mg/kg/d, i.p.) from day 1 to 5, from day 8 to 12 and days 15 to 16. On day 17, animals were submitted to the behavioral test and received pharmacological treatment. Vincristine-intoxicated rats were randomly distributed in 5 experimental groups (11 rats per group): 1. 20 vincristine/vehicle, i.p.; 2. vincristine/SPM 927 (3 mg/kg), i.p.; 3. vincristine/SPM 927 (10 mg/kg), i.p; 4. vincristine/SPM 927 (30 mg/kg), i.p.; 5. vincristine/morphine (3mg/kg), s.c. SPM 927 and morphine were respectively injected 30 and 45 min prior to the implementation of behavioral tests. 25 Cold bath test (thermal allodynia) Animals were placed on ice platform submerged approximately 1 cm below the surface of cold water (40C), such that the hairy and glabrous skin of the animal feet was in contact with the cold water. The latency before the first 30 reaction (licking, moving the paws, little leaps) was recorded with a cut off time of 30 s.
WO 2006/021412 PCT/EP2005/009067 - 36 Hot plate test (thermal allodynia/hyperaliesia) Animals were placed into a glass cylinder on a hot plate (Bioblock, France) adjusted to 38*C or 52 0 C. The latency of the first reaction (licking, moving the paws, little leaps or a jump to escape the heat) was recorded. with a cut 5 off time of 30 s. Von Frey hair stimulation test (mechanical allodynia) Rats were placed on a metallic grid floor. The nociceptive testing was done by inserting the von Frey filament (Bioseb, France) through the grid floor and 10 applying it to the plantar surface of the hind paw. A trial consisted of several applications of the different von Frey filaments (at a frequency of 1-1.5 s). The von Frey filaments were applied from filament 10 g to 100 g. The mechanical allodynia threshold was recorded as soon as the animal removed its hind paw the test was stopped and the filament number was 15 recorded. Paw pressure test (mechanical hyperalgesia) The nociceptive flexion reflex was quantified using the Randall-Selitto paw pressure device (Bioseb, France), which applies a linearly increasing 20 mechanical force to the dorsum of the rat's hind paw. The mechanical nociceptive threshold was defined as the force in grams at which the rat withdrew its paw. The cut off pressure was set to 250 g. Data analysis 25 ANOVA followed by post-hoc analysis (Dunnett's test) was used to compare groups of behavioral data in each individual time points. Nucleoside-induced pain Animals, ddC intoxication and experimental groups 30 For this study, 50 male Sprague Dawley (-220 g) rats were used (Janvier, WO 2006/021412 PCT/EP2005/009067 -37 Le Genest-St-Isle, France). Rats were group-housed (3 animals per cage) and maintained in a room with controlled temperature (21-22*C) and a reversed light-dark cycle (12h/12h) with food and water available ad libitum. All experiments were carried out in accordance with institutional guidelines. 5 Intoxication was achieved by a single injection of ddC (50 mg/kg, I.V. in tail vein). On day 10 and day 20, animals were submitted to the behavioral test and 'received pharmacological treatment. ddC-intoxicated rats were randomly distributed in 5 experimental groups (10 rats per group): 1. control/vehicle,ip, 2. ddC/vehicle, i.p.; 3. ddC/SPM 927 (3 mg/kg), i.p.; 3. 10 ddC/SPM 927 (10 mg/kg), i.p; 4. ddC/SPM 927 (30 mg/kg), i.p.; 5. ddC/morphine (3mg/kg), s.c. SPM 927 and morphine were respectively injected 30 and 45 min prior to the implementation of behavioral tests. Brushing test day 20 15 The hair on the legs, flanks, and lower back was sequentially brushed with a cotton-tipped applicator using an oscillating motion (rate of 1-2/s; 30 sec). Brushing was done with no more force than required to move the applicator through the hair such that only the pelage is disturbed. Vocalization and moderate effects to avoid the brushing were counted. 20 Results Bone cancer rat model Mechanical allodynia Fig. 1 depicts group responses to the Von Frey filaments following baseline 25 testing and following drug treatment. The statistical analysis performed using an overall 2-way ANOVA was significantly different for treatment group (p<0.01), but not for baseline vs. treatment effects. One-way ANOVA tests comparing the "cells only" group following "treatment" with each of the treatment groups revealed significant differences in the level of mechanical 30 allodynia for the morphine group (p<0.01), and the 20 and 40 mg/kg SPM 927 group (p<0.05). The 5 mg/kg morphine treatment completely reversed the allodynia that was revealed at the time of baseline testing. In addition, a highly significant difference was demonstrated between the "post-treatment" WO 2006/021412 PCT/EP2005/009067 -38 data for the healthy and tumor-injected "cells only" group (p<0.01). Statistical differences also exist between the baseline values and post-dose values for the "cells only" group and the morphine group (p<0.01). 5 Thermal hyperalgesia The data for thermal paw testing (Fig. 2) on Days 14 and 15 show that the baseline latencies to remove their paws for all tumor-injected groups were. This was confirmed to be highly significant with a two-way ANOVA, in which the drug effect was p<0.0001 and the effect of treatment was significant at 10 p<0.05. The baseline or pre-treatment data were significantly different for the "no cells" vs. "cells only" groups (Dunnett's post-hoc test; p<0.001) with the other groups showing no differences from one another. In addition, the post-treatment groups were different for "no cells" vs. "cells only" (Dunnett's; p<0.001) and "cells only" vs. SPM 927 30 mg/kg dose (Dunnett's; p<0.001). 15 The morphine control group did not show statistical significance although there was a trend indicating that morphine increased the latency to paw removal. Mechnical Hyperalqesia 20 The graph (Figure 3) shows an overall 2-way ANOVA showed significant group differences, 0<0.001. Since weight-bearing differences were defined as the weight borne by the contralateral leg minus the weight borne by the ipsilateral (tumor-injected) leg, a higher number would indicate more weight on the non-affected paw with 0 meaning equal weight distribution on both 25 paws. The baseline data collected showed a positive number, revealing that these animals are all placing more weight on their non-affected paws. In comparison to the "cells only" group, the morphine group and the 40 mg/kg SPM 927 groups showed a significant reduction (p<0.05) in the amount of weight placed on their contralateral paws following treatment, as compared 30 to the "cells only" group. Vincristine-induced pain Cold bath test WO 2006/021412 PCT/EP2005/009067 - 39 As shown in figure 4, a significant statistical difference was seen between the 6 groups (p < 0.05, Anova test). Vincristine-animals treated with the vehicle displayed very short threshold latency in the cold bath test (about 9 s) in contrast with control animals showing a time score about 14 s. 5 Treatment of vincristine-animals with SPM 927 induced a significant increase (p < 0.05, Dunnett's test) in the threshold latency, which became indeed comparable to that of control animals, especially for the treatment doses of 10 and 30 mg/kg. At 3 mg/kg however, the threshold latency was slightly greater than that of vincristine-animals, although statistical difference 10 was not reached. Morphine treatment extended the threshold latency of vincristine-animals to a level far above that obtained from control rats. Hot plate test at 38 0C Figure 5 shows that the threshold latency of vincristine-animals in the hot 15 plate (38 *C) test was significantly shorter than that of control animals (p < 0.05, Dunnett's test). Treatment of vincristine-rats with SPM 927 at 3, 10 and 30 mg/kg induced a significant (p < 0.05, Dunnett's test) increase in the threshold latency. With the treatment doses of 10 and 30 mg/kg, the performance of STZ-rats became comparable with that of control animals. 20 Similarly to SPM 927 at 10 and 30 mg/kg, morphine at 3 mg/kg extended the threshold latency of STZ-animals to a level comparable with that of control rats. Hot plate test at 52 *C 25 As illustrated in figure 6, the paw withdrawal latency of vincristine-rats was significantly shorter than that of control animals (p < 0.05, Dunnett's test). Treatment of vincristine-rats with SPM 927 induced a significant increase (p < 0.05, Dunnett's test) in paw withdrawal latency in as compared with vehicle-treated animals. The effect obtained with the doses of 3 and 10, 30 30 mg/kg was comparable with that of control. Paw pressure test Using the analgesy-meter of Randall & Selitto, vincristine-animals WO 2006/021412 PCT/EP2005/009067 -40 demonstrated a marked decrease in the paw withdrawal latency as compared with the performance of control animals (figure 7). Treatment of vincristine-rats with SPM 927 at the doses of 10 and 30 mg/kg, but not at 3 mg/kg, induced a significant increase (p < 0.05, Dunnett's test) in paw 5 withdrawal latency of vincristine-rats. In this test, morphine treatment did not modify the performance of vincristine-rats (p > 0.05, Dunnett's test). Von Frey Filament test In this test. (figure 8), the paw withdrawal latency of vincristine-rats was 10 significantly reduced (about 20 g) as compared with that of control rats (about 60 g). Treatment with SPM 927 extended the paw withdrawal latency of vincristine rats. The difference with the vehicle-treated group reached the significance level with the treatment doses of 10 and 30 mg/kg (p < 0.05, Dunnett's test). Morphine treatment restored the performance of vincristine 15 rats to a level comparable with that of control group. Nucleoside-induced pain rat model Thermal allodynia As shown in figure 9, a significant statistical difference was seen between 20 the 6 groups (p < 0.05, Anova test). ddC-animals treated with the vehicle displayed very short threshold latency in the cold bath test (about 11 s) in contrast with control animals showing a time score about 20 s. Treatment of ddC-animals with SPM 927 induced a significant increase (p < 0.05, Dunnett's test) in the threshold latency, which became indeed comparable to 25 that of control animals for the 3 test doses, 3, 10 and 30 mg/kg. Morphine treatment extended the threshold latency of ddC-animals to a level comparable to that obtained from control rats. Brushing test at day 20 30 Figure 10 shows the results of brushing test performed on D 20. The animals treated with SPM 927 (at 3, 10 and 30 mg/kg) displayed a significant decrease in the total number of cries (p < 0.05, Dunnett's test). Here again, WO 2006/021412 PCT/EP2005/009067 -41 morphine at 3 mg/kg was able to significantly decrease the total number of cries in the ddC-animals. Von Frey filament test at D10 5 In this test (figure 11), the paw withdrawal latency of ddC-rats was significantly reduced (about 50 g) as compared with that of control rats (about 85 g). Treatment with SPM 927 extended the paw withdrawal latency of ddC rats. The difference with the vehicle-treated group reached the significance level with the treatment doses of 3, 10 and 30 mg/kg (p < 0.05, 10 Dunnett's test) to a level comparable to control group. Morphine treatment at 3 mg/kg restored the performance of ddC-rats to a level similar to control group. Hot plate test at 52 *C at D20 15 As illustrated in figure 12, the paw withdrawal latency of ddC-rats was significantly shorter than that of control animals (p < 0.05, Dunnett's test). Treatment of ddC-rats with SPM 927 only at the dose of 30 mg/kg, induced a significant increase (p < 0.05, Dunnett's test) in paw withdrawal latency in as compared with vehicle-treated animals. The effect obtained with the doses 20 of 3 and 10 mg/kg was comparable with untreated vehicle group. Following morphine treatment, the performance of ddC-rats became similar with that of control animals. Paw pressure test at D10 25 Using the analgesy-meter of Randall & Selitto, ddC-animals demonstrated a marked decrease in the paw withdrawal latency as compared with the performance of control animals (figure 13). Treatment of ddC-rats with SPM 927 at all the 3 doses of 3, 10and 30 mg/kg induced a significant increase (p < 0.05, Dunnett's test) in paw withdrawal latency of ddC-rats. Again 30 morphine treatment increased the performance of ddC-rats (p < 0.05, Dunnett's test).
WO 2006/021412 PCT/EP2005/009067 -42 Conclusion Systemic SPM 927 produced a dose-dependent anti-allodynic and anti hyperalgeisc effect in a rat model of bone cancer pain, chemotherapy- and nucleoside-induced pain following single dose administration. Thus, SPM 927 and related compounds as disclosed in formulae (Ib) or/and (11b) are useful for the treatment of pain during cancer, e.g. bone cancer pain, after treatment with chemotherapy and nucleosides in humans.
WO 2006/021412 PCT/EP2005/009067 -43 References Medhurst, S.J., Walker, K., Bowes, M., Kidd, B.L., Glatt, M., Muller, M., Hattenberger, M., Vaxelaire, J., O'Reilly, T.O., Wotherspoon, G., Winter, J., 5 Green, J., and Urban, L. A rat model of bone cancer pain. Pain, 96: 129 140, 2002. Casey EB, Jellife AM, Le Quesne PM, Millett YL. Vincristine neuropathy. Clinical and electrophysiological observations. Brain 1973; 96: 69-86. 10 Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of commonly used antineoplastic agents (second of two parts). N Engl J Med. 1974 ; 291(3): 127-33. Quasthoff S, Hartung HP Chemotherapy-induced peripheral neuropathy J 15 Neurol (2002) 249 : 9-17. Kimberly D. Tanner, David B. Reichling, and Jon D. Levine. Nociceptor Hyper-Responsiveness during Vincristine-Induced Painful Peripheral Neuropathy in the Rat. J Neurosc (1998) 18(16):6480-6491. 20 Kaplan RS, Wiernik PH Neurotoxicity of antineoplastic drugs. (1982) Semin Oncol 9:103-130. Owellen RJ, Hartke CA, Dickerson RM, Hains FO (1976) Inhibition of tubulin-microtubule polymerization by drugs of the vinca alkaloid class. 25 Cancer Res 36:1499 -1502. Sander SG, Tobin W, Henderson ES (1969) Vincristine-induced neuropathy. A clinical study of fifty leukemic patients. Neurology 19:367-374. Holland JF, Scharlau C, Gailani S, Krant MJ, Olson KB, Shnider BI, Lynch 30 JJ, Owens A, Carbone PP, Colsky J, Grob SP, Hall TC (1973) Vincristine treatment of advanced cancer: cooperative study of 392 cases. Cancer Res 33:1258 -1264.
WO 2006/021412 PCT/EP2005/009067 -44 McCarthy GM, Skillings JR (1992) Jaw and other orofacial pain in patients receiving vincristine for the treatment of cancer. Oral Surg Oral Med Oral Pathol 74:299 -304. 5 Aley KO, Levine JD. Different mechanisms mediate development and expression of tolerance and dependence for peripheral mu-opioid antinociception in rat. J Neurosci 1997;17:8018-23. Aley KO, Levine JD. Role of protein kinase A in the maintenance of 10 inflammatory pain. J Neurosci 1999;19:2181-6. Aley KO, Levine JD. Different peripheral mechanisms mediate enhanced nociception in metabolic/toxic and traumatic painful peripheral neuropathies in the rat. Neuroscience 2002;111:389-97. 15 Aley KO, McCarter G, Levine JD. Nitric oxide signaling in pain and nociceptor sensitization in the rat. J Neurosci 1998;18:7008 Kaplan RS, Wiernik PH Neurotoxicity of antineoplastic drugs. (1982) Semin Oncol 9:103-130. 20 Cohen J. Therapies. Confronting the limits of success. Science 2002;296: 2320-4. Dalakas MC. Peripheral neuropathy and antiretroviral drugs. J Peripher Nerv 25 Syst 2001;6:14-20. Dalakas MC, Semino-Mora C, Leon-Monzon M. Mitochondrial alterations with mitochondrial DNA depletion in the nerves of AIDS patients with peripheral neuropathy induced by 2030-dideoxycytidine (ddC). Lab Invest so 2001;81:1537-44. Dina OA, Barletta J, Chen X, Mutero A, Martin A, Messing RO, Levine JD. Key role for the epsilon isoform of protein kinase C in painful alcoholic P \OPER\DA)H\Speci30185203 vol a.ed 156 doc-2M 36 07 - 45 neuropathy in the rat. J Neurosci 2000;20:8614-9. Dubinsky RM, Yarchoan R, Dalakas M, Broder S. Reversible axonal neuropathy from the treatment of AIDS and related disorders with 20,30-dideoxycytidine 5 (ddC). Muscle Nerve 1989;12:856-60 Josepha EK, Chena X, Khasara SG, Levinea JD. Novel mechanism of enhanced nociception in a model of AIDS therapy-induced painful peripheral neuropathy in the rat. Pain 107 (2004) 147-158. 10 Williams D, Geraci A, Simpson DM. AIDS and AIDS-treatment neuropathies. Curr Neurol Neurosci Rep 2001;1:533-8. Yatvin MB, Li W, Meredith MJ, Shenoy MA. Improved uptake and retention of 15 lipophilic prodrug to improve treatment of HIV. Adv Drug Deliv Rev 1999;39:165 8. Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and 20 "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. The reference in this specification to any prior publication (or information derived 25 from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

Claims (26)

1. Use of a compound having the Formula (1lb) 5 H H H Ar-C Hr-N-C-C-N-C-R1 Formula (lIb) 10 wherein Ar is phenyl which is unsubstituted or substituted with at least one halo group, 15 R 3 is CH
2 -Q, wherein Q is lower alkoxy containing 1-3 carbon atoms, and R 1 is lower alkyl containing 1-3 carbon atoms or of a pharmaceutically acceptable salt thereof, 20 for the preparation of a pharmaceutical composition for the prevention, alleviation or/and treatment of tumor pain, chemotherapy induced pain, or/and pain induced by at least one nucleoside or/and at least one nucleoside analogue. 25 2. Use according to claim 1, wherein the tumor pain is tumor pain associated with AIDS, bone cancer pain, pain produced during tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves, metastasis-induced pain, metastasis-induced bone cancer pain or/and pain induced by metastatic disease in patients with breast, prostate or 30 lung cancer, wherein the chemotherapy-induced pain is chemotherapy- C:\NRPonbl\DCCUX'f\329659_. DOC-1 11112010 -47 induced neuropathic pain, vinca alkaloid-induced pain or/and vincristine induced pain, or/and wherein the nucleoside or/and nucleoside analogue induced pain is painful peripheral neuropathy induced by nucleosides or/and nucleoside analogues, pain induced by anti-tumor or/and anti-viral nucleoside 5 analogues, pain induced by anti-viral nucleoside analogues in AIDS therapy, or/and pain induced by AZT, ddC, ddl or/and d4T.
3. Use according to claim 1 or 2 wherein Ar is unsubstituted phenyl. 10
4. Use according to claim 1 or 2 wherein halo is fluoro.
5. Use according to any one of claims 1 to 4 wherein R 3 is CH 2 -Q, wherein Q is methoxy. 15
6. Use according to claim 1 or 2 wherein the compound is (R)-2-acetamido-N-benzyl-3-methoxy-propionamide; O-methyl-N-acetyl-D-serine-m-fluorobenzylamide; or O-methyl-N-acetyl-D-serine-p-fluorobenzylamide. 20
7. Use according to claim 1 or 2 wherein the compound is in the R configuration and has the formula H R-NH-C-C-N-C-R1 25 III o R 3 R wherein 30 R is benzyl which is unsubstituted or substituted with at least one halo C:\NRPonbl\DCC!JX-'3298659 I DC- I/1ll2010 -48 group; R 2 is hydrogen, 5 R 3 is CH 2 -Q, wherein Q is lower alkoxy containing 1-3 carbon atoms, and R 1 is methyl; or a pharmaceutically acceptable salt thereof. 10
8. Use according to claim 7, wherein the compound is substantially enantiopure.
9. Use according to claim 7 or 8 wherein R is unsubstituted benzyl. 15
10. Use according to claim 7 or 8 wherein halo is fluoro.
11. Use according to any one of claims 7 to 10 wherein R 3 is CH 2 -Q, wherein Q is methoxy. 20
12. Use according to claim 1 or 2, wherein the compound of Formula (lib) is (R)-2-Acetamido-N-benzyl-3-methoxypropionamide or a pharmaceutically acceptable salt thereof.
13. Use according to claim 12 wherein the compound is substantially 25 enantiopure.
14. Use according to any one of claims 1 to 13, wherein the pharmaceutical composition is prepared for treatment with doses of the compound at least of 100 mg/day, preferably at least of 200 30 mg/day, more preferably at least of 300 mg/day, most preferably at C:\NRPonbl\DCC\JX-l\299659 DOC.C-1111/2010 - 49 least of 400 mg/day.
15. Use according to any one of claims 1 to 14, wherein the pharmaceutical composition is prepared for treatment with doses of 5 the compound at a maximum of 6 g/day, more preferably at a maximum of 1 g/day and most preferably at a maximum of 600 mg/day.
16. Use according to any one of claims 1 to 15, wherein the 10 pharmaceutical composition is prepared for treatment with increasing daily doses until a predetermined daily dose is reached which is maintained during the further treatment.
17. Use according to any one of claims 1 to 16, wherein the 15 pharmaceutical composition is prepared for treatment in three doses per day, preferably two doses per day, more preferably in a single dose per day.
18. Use according to any one of claims 1 to 17, wherein the 20 pharmaceutical composition is prepared for an administration resulting in a plasma concentration of 0.1 to 15 pg/ml (trough) and 5 to 18.5 pg/ml (peak), calculated as an average over a plurality of treated subjects. 25
19. Use according to any one of claims 1 to 18, wherein the pharmaceutical composition is prepared for oral or i.v. administration.
20. Use according to any one of claims 1 to 19, wherein the pharmaceutical composition further comprises an active agent for the 30 prevention, alleviation, or/and treatment of a viral infection such as C:NRPonb\WCCX-l\U902gSIl.DOC-23'2/2011 - 50 HIV infection including AIDS, of cancer such as breast cancer, prostate cancer, lung cancer, bone cancer, metastatic disease, or/and of tumor progression by infiltration in or pressure on bone, viscera, soft tissue or nerves, wherein said further active agent is selected from the anti 5 retroviral nucleoside analogue AIDS therapy drugs ddC (2',3' dideoxycytidine), ddl (2',3'-dideoxyinosine) or d4T (2',3'-didehydro-3' deoxythymidine), vinca alkaloids, taxol, suramin, cisplatin, carboplatin and oxaliplatin. 10
21. Use according to claim 20 wherein the pharmaceutical composition comprises a single dose form or comprises a separate dose form comprising a first composition comprising a compound as defined in any one of claims 1 and 3 to 13 and a second composition comprising the further active agent. 15
22. Use according to any one of claims 1 to 21 wherein the pharmaceutical composition is prepared for administration in mammals and/or humans. ~ C:RvaribilCCMUM8591DOC-1llM/210 -51
23. A method for the prevention, alleviation or/and treatment of tumor pain, chemotherapy induced pain, or/and pain induced by at. least one nucleoside or/and at least one nucleoside analogue In a mammal, including a human being, comprising administering at least one compound of formula (lib) or a pharmaceutically acceptable saft thereof, H H H Ar-CHL C-1 Formula (l1b) wherein in Formula (lib), Ar is phenyl which is unsubstituted or substituted with at least one halo group, is CtHrQ, wherein Q Is lower alkoxy containing 1-3 carbon atoms, and R 1 is lower alkyl containina 1-3 carbon atoms.
24. The method according to claim 23 comprising administering a compound of Formula (tIb) or a pharmaceutcallY acceptable salt thereof in combination with a further active agent for the prevention, alleviation or/and treatment of a viral infection such as HIV Infection Including AIDS, of cancer such as breast cancer, prostate cancer, lung cancer, bone cancer, metastatic disease, or/and of tumor progression by infiltration In or pressure on bone, viscera, soft tissue or nerves, wherein said further- active agent is selected from the anti-retroviral nucleoside analogue AIDS therapy drugs ddC (2',3'-didexyCYtidIne). ddl (2',3'-dideoxyinosine) or d4T (2',3'.didehydro3-deoxythymidine). vinca alkaloids, taxol, suramin, cisplatin, carboplatin and oxalliplatin. C:\NRPorbl\DCC\JXT\14902N5_I DOC-2IA2/20 11 - 52
25. The method of claim 23 or 24, wherein the compound of Formula (11b) is (R)-2-acetamido-N-benzyl-3-methoxy-propionamide. 5
26. A use according to claim 1, or a method according to claim 23, substantially as hereinbefore described with reference to any one of the examples.
AU2005276634A 2004-08-27 2005-08-22 Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain Ceased AU2005276634B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US60481004P 2004-08-27 2004-08-27
EP04020402A EP1629839A1 (en) 2004-08-27 2004-08-27 Use of peptide compounds for treating bone cancer pain, chemotherapy- and nucleoside-induced pain
US60/604,810 2004-08-27
EP04020402.6 2004-08-27
PCT/EP2005/009067 WO2006021412A2 (en) 2004-08-27 2005-08-22 Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain

Publications (2)

Publication Number Publication Date
AU2005276634A1 AU2005276634A1 (en) 2006-03-02
AU2005276634B2 true AU2005276634B2 (en) 2011-03-24

Family

ID=35456911

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005276634A Ceased AU2005276634B2 (en) 2004-08-27 2005-08-22 Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain

Country Status (17)

Country Link
US (2) US7687553B2 (en)
EP (1) EP1781276B1 (en)
JP (1) JP2008510758A (en)
KR (1) KR20070045271A (en)
AR (1) AR050612A1 (en)
AT (1) ATE471717T1 (en)
AU (1) AU2005276634B2 (en)
BR (1) BRPI0514721A (en)
CA (1) CA2573125A1 (en)
DE (1) DE602005021970D1 (en)
EA (1) EA014055B1 (en)
IL (1) IL181383A0 (en)
MX (1) MX2007002269A (en)
NO (1) NO20071585L (en)
NZ (1) NZ552651A (en)
PL (1) PL1781276T3 (en)
WO (1) WO2006021412A2 (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19633169A1 (en) 1996-08-17 1998-02-19 Teves Gmbh Alfred Electric motor / pump unit
DK1243262T3 (en) * 2001-03-20 2006-10-02 Sanol Arznei Schwarz Gmbh Hitherto unknown use of a peptide class of compounds to treat non-neuropathic inflammatory pain
PT1243263E (en) * 2001-03-21 2003-03-31 Sanol Arznei Schwarz Gmbh NEW USE OF A PEPTIDICAL COMPOSITE CLASS FOR THE TREATMENT OF ALODINIA OR OTHER DIFFERENT TYPES OF CHRONIC OR GHOST PAIN
KR20060111524A (en) * 2003-12-02 2006-10-27 쉬바르츠파르마에이지 Novel use of peptide compounds for treating central neuropathic pain
US20070042969A1 (en) * 2004-03-26 2007-02-22 Srz Properties, Inc. Combination therapy for pain in painful diabetic neuropathy
US20100256179A1 (en) * 2004-03-26 2010-10-07 Ucb Pharma Gmbh Combination therapy for pain in painful diabetic neuropathy
MXPA06011937A (en) 2004-04-16 2007-01-26 Sanol Arznei Schwarz Gmbh Use of peptidic compounds for the prophylaxis and treatment of chronic headache.
EP1604655A1 (en) * 2004-06-09 2005-12-14 Schwarz Pharma Ag Novel use of peptide compounds for treating pain in trigeminal neuralgia
MX2007002269A (en) * 2004-08-27 2007-06-15 Sanol Arznei Schwarz Gmbh Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain.
EP1642889A1 (en) * 2004-10-02 2006-04-05 Schwarz Pharma Ag Improved synthesis scheme for lacosamide
US20060252749A1 (en) * 2005-01-28 2006-11-09 Srz Properties, Inc. Lacosamide for add-on therapy of psychosis
US20070043120A1 (en) * 2005-08-18 2007-02-22 Bettina Beyreuther Therapeutic combination for painful medical conditions
EP1754476A1 (en) * 2005-08-18 2007-02-21 Schwarz Pharma Ag Lacosamide (SPM 927) for treating myalgia, e.g. fibromyalgia
US20070048372A1 (en) * 2005-08-18 2007-03-01 Srz Properties, Inc. Method for treating non-inflammatory osteoarthritic pain
CN101466390B (en) 2006-06-15 2014-03-12 优时比制药有限公司 Pharmaceutical composition with synergistic anticonvulsant effect
EP1873527A1 (en) * 2006-06-30 2008-01-02 Schwarz Pharma Ag Method for identifying CRMP modulators
KR20150003925A (en) 2006-06-15 2015-01-09 유씨비 파르마 게엠베하 Pharmaceutical composition with synergistic anticonvulsant effect
WO2011037833A2 (en) 2009-09-23 2011-03-31 The University Of North Carolina At Chapel Hill Novel n-benzylamide substituted derivatives of 2-(acylamido)acetic acid and 2-(acylamido)propionic acids: potent neurological agents
US20130251813A1 (en) 2010-12-02 2013-09-26 Ucb Pharma Gmbh Formulation of lacosamide
EP2468261A1 (en) 2010-12-02 2012-06-27 UCB Pharma GmbH Formulation of lacosamide
US9134395B2 (en) 2012-03-07 2015-09-15 Freescale Semiconductor, Inc. Method for testing comparator and device therefor
EP3402017A4 (en) * 2016-01-07 2019-01-16 Panasonic Intellectual Property Management Co., Ltd. Light-emitting device

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002015922A2 (en) * 2000-08-25 2002-02-28 Research Corporation Technologies, Inc. New uses for amino acid anticonvulsants for treatment of pain
WO2002074297A1 (en) * 2001-03-21 2002-09-26 Schwarz Pharma Ag Novel use of a peptide class of compound for treating allodynia or other different types of chronic or phantom pain

Family Cites Families (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2937698A1 (en) 1979-09-18 1981-04-02 A. Nattermann & Cie GmbH, 5000 Köln N-PROPIONYLSARCOSINANILIDES, THEIR PRODUCTION METHOD AND MEDICINAL PRODUCTS BASED ON THEM
FR2480747A1 (en) 1980-04-17 1981-10-23 Roques Bernard AMINO ACID DERIVATIVES AND THEIR THERAPEUTIC APPLICATION
US4533657A (en) 1981-07-24 1985-08-06 Sterling Drug Inc. Analgesic dipeptide amides and method of use and composition thereof
FR2518088B1 (en) 1981-12-16 1987-11-27 Roques Bernard NOVEL AMINO ACID DERIVATIVES AND THEIR THERAPEUTIC APPLICATION
US4510082A (en) 1983-03-07 1985-04-09 Eli Lilly And Company Pharmacologically active peptides
JPH0680079B2 (en) 1984-11-09 1994-10-12 エーザイ株式会社 Polypeptide
US5378729A (en) 1985-02-15 1995-01-03 Research Corporation Technologies, Inc. Amino acid derivative anticonvulsant
GB9103454D0 (en) 1991-02-19 1991-04-03 Pfizer Ltd Therapeutic agents
US5656267A (en) 1991-08-22 1997-08-12 Sagen; Jacqueline Implantable cells that alleviate chronic pain in humans
TW217417B (en) 1991-12-04 1993-12-11 Manyu Seiyaku Kk
US5585358A (en) * 1993-07-06 1996-12-17 Yissum Research Development Corporation Of The Hebrew University Of Jerusalem Derivatives of valproic acid amides and 2-valproenoic acid amides, method of making and use thereof as anticonvulsant agents
GB9504854D0 (en) 1994-03-31 1995-04-26 Zeneca Ltd Nitrogen derivatives
US5536853A (en) 1994-04-11 1996-07-16 Chiron Corporation Opiate receptor ligands
MY113062A (en) 1994-05-10 2001-11-30 The Wellcome Foundation Ltd Amide derivatives and their therapeutic use
US5508266A (en) 1994-06-22 1996-04-16 Ciba-Geigy Corporation Gem-disubstituted amino acid derivatives
HUT77282A (en) 1994-10-05 1998-03-30 Darwin Discovery Limited Peptidyl compounds and their therapeutic use as inhibitors of metalloproteases
DE19533023B4 (en) 1994-10-14 2007-05-16 Basf Ag New carboxylic acid derivatives, their preparation and use
US5780589A (en) 1994-11-30 1998-07-14 The United States Of America As Represented By The Department Of Health And Human Services Ultraselective opioidmimetic peptides and pharmacological and therapeutic uses thereof
US5760038A (en) 1995-02-06 1998-06-02 Bristol-Myers Squibb Company Substituted biphenyl sulfonamide endothelin antagonists
US5849737A (en) * 1995-04-14 1998-12-15 The Regents Of The University Of California Compositions and methods for treating pain
JP4313435B2 (en) 1995-07-24 2009-08-12 トラスティーズ オブ ボストン ユニバーシティー Inhibition of NMDA receptor activity by pregnenolone sulfate derivatives
US6114390A (en) 1995-11-30 2000-09-05 Karl Thomae Gmbh Amino acid derivatives, pharmaceutical compositions containing these compounds and processes for preparing them
US6037324A (en) 1996-01-04 2000-03-14 Leukosite, Inc. Inhibitors of MAdCAM-1-mediated interactions and methods of use therefor
GB9601724D0 (en) 1996-01-29 1996-03-27 Merck Sharp & Dohme Therapeutic agents
US5773475A (en) * 1997-03-17 1998-06-30 Research Corporation Technologies, Inc. Anticonvulsant enantiomeric amino acid derivatives
DE19614542A1 (en) 1996-04-12 1997-10-16 Basf Ag New carboxylic acid derivatives, their production and use
DE19614534A1 (en) 1996-04-12 1997-10-16 Basf Ag New carboxylic acid derivatives, their production and use
DE19614533A1 (en) 1996-04-12 1997-10-16 Basf Ag New alpha-hydroxy acid derivatives, their production and use
US5773455A (en) 1996-06-28 1998-06-30 Biomeasure, Incorporated Inhibitors of prenyl transferases
US6277825B1 (en) 1996-07-22 2001-08-21 University Of Utah Research Foundation Use of conantokins for treating pain
NZ332762A (en) 1996-07-24 2000-09-29 Warner Lambert Co Isobutylgaba and its derivatives for the treatment of pain
US6589994B1 (en) 1996-08-30 2003-07-08 Nps Pharmaceuticals, Inc. Treating a variety of pathological conditions, including spasticity and convulsions, by effecting a modulation of CNS activity with isovaleramide, isovaleric acid, or a related compound
US6126939A (en) 1996-09-03 2000-10-03 Yeda Research And Development Co. Ltd. Anti-inflammatory dipeptide and pharmaceutical composition thereof
DE19636046A1 (en) 1996-09-05 1998-03-12 Basf Ag New carboxylic acid derivatives, their production and use as mixed ET¶A¶ / ET¶B¶ receptor antagonists
US5866585A (en) 1997-05-22 1999-02-02 Synchroneuron, Llc Methods of treating tardive dyskinesia using NMDA receptor antagonists
AU7937298A (en) 1997-07-08 1999-02-08 Ono Pharmaceutical Co. Ltd. Amino acid derivatives
US6737408B1 (en) 1997-08-07 2004-05-18 University Of Cincinnati Compounds for control of appetite, blood pressure, cardiovascular response, libido, and circadian rhythm
WO1999007413A1 (en) 1997-08-11 1999-02-18 Algos Pharmaceutical Corporation Substance p inhibitors in combination with nmda-blockers for treating pain
DE19743143A1 (en) 1997-09-30 1999-04-01 Knoll Ag Combination pharmaceutical preparations
CN1278251A (en) 1997-10-31 2000-12-27 Basf公司 Novel carboxylic acid derivatives which carry amide side chains, prodn. of same, and use as endothelin receptor antagonists
US6028102A (en) 1998-02-24 2000-02-22 Yissum Research Development Company Of The Hebrew University Of Jerusalem Anticonvulsant drugs and pharmaceutical compositions thereof
IL143935A (en) 1999-09-03 2005-08-31 Actelion Pharmaceuticals Ltd Bis-sulfonamides
SE0001373D0 (en) 2000-04-13 2000-04-13 Karolinska Innovations Ab NPY Y1 receptor agonists and antagonists
US20020052418A1 (en) * 2000-08-17 2002-05-02 Mitchell Shirvan Use of derivatives of valproic acid amides and 2-valproenic acid amides for the treatment or prevention of pain and/or headache disorders
DE60134337D1 (en) 2000-11-21 2008-07-17 Ucb Pharma Sa N-ALKYLATED GABA COMPOUNDS, METHODS THEREOF
DK1243262T3 (en) * 2001-03-20 2006-10-02 Sanol Arznei Schwarz Gmbh Hitherto unknown use of a peptide class of compounds to treat non-neuropathic inflammatory pain
NZ527276A (en) 2001-03-27 2004-11-26 Actelion Pharmaceuticals Ltd 1,2,3,4-tetrahydroisoquinoline derivatives as urotensin II receptor antagonists
BR0209155A (en) 2001-04-26 2004-10-05 Bristol Myers Squibb Co Pharmaceutical tablet having a high api content
ITMI20011308A1 (en) 2001-06-21 2002-12-21 Nicox Sa DRUGS FOR CHRONIC PAIN
AU2003243180A1 (en) 2002-05-17 2003-12-12 Xenoport, Inc. Amino acid conjugates providing for sustained systemic concentrations of gaba analogues
US7384934B2 (en) 2002-08-05 2008-06-10 Eli Lilly And Company Piperazine substituted aryl benzodiazepines
EP1575919A1 (en) 2002-11-11 2005-09-21 Bayer HealthCare AG Phenyl or heteroaryl amino alkane derivatives as ip receptor antagonist
WO2004046178A2 (en) 2002-11-18 2004-06-03 Stewart John M Shrew paralytic peptide for use in neuromuscular therapy
US7687080B2 (en) * 2002-11-25 2010-03-30 Taraxos Inc. Treatment of neuropathy
ATE353633T1 (en) 2002-12-19 2007-03-15 Pharmacia Corp NON-HYGROSCOPIC FORMULATION CONTAINING A HYGROSCOPIC ACTIVE INGREDIENT
EP1589959A2 (en) 2003-01-30 2005-11-02 Dynogen Pharmaceuticals Inc. Methods of treating lower urinary tract disorders using sodium channel modulators
CA2514574A1 (en) 2003-01-30 2004-08-12 Dynogen Pharmaceuticals, Inc. Use of sodium channel modulators for treating gastrointestinal tract disorders
US20040265375A1 (en) 2003-04-16 2004-12-30 Platteeuw Johannes J. Orally disintegrating tablets
US20040266743A1 (en) 2003-05-09 2004-12-30 Pharmacia Corporation Combination of an aldosterone receptor antagonist and a renin inhibitor
US7320675B2 (en) 2003-08-21 2008-01-22 Cardiac Pacemakers, Inc. Method and apparatus for modulating cellular metabolism during post-ischemia or heart failure
KR20060111524A (en) * 2003-12-02 2006-10-27 쉬바르츠파르마에이지 Novel use of peptide compounds for treating central neuropathic pain
EP1537862A1 (en) 2003-12-02 2005-06-08 Schwarz Pharma Ag Novel use of peptide compounds for treating central neuropathic pain
US20060009384A1 (en) 2003-12-05 2006-01-12 David Rudd Novel use of peptide compounds for treating status epilepticus or related conditions
EP1541138A1 (en) 2003-12-05 2005-06-15 Schwarz Pharma Ag Novel use of peptide compounds for treating status epilepticus or related conditions
EP1579858A1 (en) 2004-03-26 2005-09-28 Schwarz Pharma Ag Novel use of peptide compounds for treating pain in painful diabetic neuropathy
US20100256179A1 (en) * 2004-03-26 2010-10-07 Ucb Pharma Gmbh Combination therapy for pain in painful diabetic neuropathy
US20070042969A1 (en) * 2004-03-26 2007-02-22 Srz Properties, Inc. Combination therapy for pain in painful diabetic neuropathy
US20050227961A1 (en) 2004-04-08 2005-10-13 Vela Pharmaceuticals, Inc. Compositions and methods for treatment of neuropathic pain, fibromyalgia and chronic fatigue syndrome
MXPA06011937A (en) * 2004-04-16 2007-01-26 Sanol Arznei Schwarz Gmbh Use of peptidic compounds for the prophylaxis and treatment of chronic headache.
EP1604654A1 (en) 2004-05-18 2005-12-14 Schwarz Pharma Ag Novel use of peptide compounds for treating dyskinesia
EP1604656A1 (en) * 2004-06-09 2005-12-14 Schwarz Pharma Ag Novel use of peptide compounds for treating amyotrophic lateral sclerosis (ALS)
EP1604655A1 (en) 2004-06-09 2005-12-14 Schwarz Pharma Ag Novel use of peptide compounds for treating pain in trigeminal neuralgia
US7427601B2 (en) 2004-06-24 2008-09-23 Schwarz Pharma Ag Method for treating tremor
MX2007002269A (en) 2004-08-27 2007-06-15 Sanol Arznei Schwarz Gmbh Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain.
EP1642889A1 (en) * 2004-10-02 2006-04-05 Schwarz Pharma Ag Improved synthesis scheme for lacosamide
US20060252749A1 (en) * 2005-01-28 2006-11-09 Srz Properties, Inc. Lacosamide for add-on therapy of psychosis
EP1688137A1 (en) 2005-01-28 2006-08-09 Schwarz Pharma Ag SPM 927 for add-on therapy of schizophrenia
EP1754476A1 (en) * 2005-08-18 2007-02-21 Schwarz Pharma Ag Lacosamide (SPM 927) for treating myalgia, e.g. fibromyalgia
US20070043120A1 (en) * 2005-08-18 2007-02-22 Bettina Beyreuther Therapeutic combination for painful medical conditions
US20070048372A1 (en) * 2005-08-18 2007-03-01 Srz Properties, Inc. Method for treating non-inflammatory osteoarthritic pain
CN101466390B (en) * 2006-06-15 2014-03-12 优时比制药有限公司 Pharmaceutical composition with synergistic anticonvulsant effect
KR20150003925A (en) * 2006-06-15 2015-01-09 유씨비 파르마 게엠베하 Pharmaceutical composition with synergistic anticonvulsant effect
EP1873527A1 (en) * 2006-06-30 2008-01-02 Schwarz Pharma Ag Method for identifying CRMP modulators
WO2009053070A1 (en) * 2007-10-23 2009-04-30 Schwarz Pharma Ag Compounds for treating demyelination conditions

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002015922A2 (en) * 2000-08-25 2002-02-28 Research Corporation Technologies, Inc. New uses for amino acid anticonvulsants for treatment of pain
WO2002074297A1 (en) * 2001-03-21 2002-09-26 Schwarz Pharma Ag Novel use of a peptide class of compound for treating allodynia or other different types of chronic or phantom pain

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HOVINGA C A: Idrugs 01 MAY 2003 UK, vol. 6, no. 5, 1 May 2003, pages 479-485 *
MCCLEANE G et al: Neuroscience Letters, vol. 352, no. 2,4 December 2003, pages 117-120 *
NICOLAUS B J R: Decision Making In Drug Research, XX, XX, 1983, pages 173-186 *
Quasthoff S. et al, J Neurol. 2002 Jan; 249(1): 9-17 *

Also Published As

Publication number Publication date
NZ552651A (en) 2010-07-30
EP1781276B1 (en) 2010-06-23
PL1781276T3 (en) 2010-11-30
MX2007002269A (en) 2007-06-15
ATE471717T1 (en) 2010-07-15
KR20070045271A (en) 2007-05-02
US7687553B2 (en) 2010-03-30
DE602005021970D1 (en) 2010-08-05
EA200700493A1 (en) 2007-08-31
US8536137B2 (en) 2013-09-17
AU2005276634A1 (en) 2006-03-02
US20060046957A1 (en) 2006-03-02
CA2573125A1 (en) 2006-03-02
WO2006021412A2 (en) 2006-03-02
JP2008510758A (en) 2008-04-10
NO20071585L (en) 2007-04-30
US20100029543A1 (en) 2010-02-04
AR050612A1 (en) 2006-11-08
BRPI0514721A (en) 2008-06-24
WO2006021412A3 (en) 2006-04-27
EA014055B1 (en) 2010-08-30
IL181383A0 (en) 2008-03-20
EP1781276A2 (en) 2007-05-09

Similar Documents

Publication Publication Date Title
AU2005276634B2 (en) Use of peptide compounds for treating bone cancer pain, chemotherapy-and nucleoside-induced pain
AU2005251465B2 (en) Novel use of peptide compounds for treating pain in trigeminal neuralgia
US7718161B2 (en) Method for treating a motoneuron disorder
AU2005226928B2 (en) Novel use of peptide compounds for treating pain in painful diabetic neuropathy
CA2651684C (en) Peptide compounds for treating refractory status epilepticus
US20090018197A1 (en) Methods for treating status epilepticus and related conditions
AU2004294714B2 (en) Novel use of peptide compounds for treating central neuropathic pain
EP1753414A1 (en) Novel use of peptide compounds for treating dyskinesia
KR20040004369A (en) Novel use of a peptide class of compound for treating non neuropathic inflammatory pain
EP1537862A1 (en) Novel use of peptide compounds for treating central neuropathic pain
EP1629839A1 (en) Use of peptide compounds for treating bone cancer pain, chemotherapy- and nucleoside-induced pain
ES2348040T3 (en) USE OF PEPTIDIC COMPOUNDS TO TREAT PAIN RELATED TO BONE CANCER AND PAIN INDUCED BY CHEMOTHERAPY AND NUCLEOSIS.
KR20070018067A (en) Novel use of peptide compounds for treating pain in trigeminal neuralgia

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired